Buscar en esta Enciclopedia

jueves, 25 de octubre de 2012

RECAÍDAS LOCALES POSTERIOR A TRATAMIENTO RADICAL EVALUACIÓN DE INFILTRACIÓN DEL COMPLEJO AREOLA-PEZÓN


Rev Venez Oncol
Recaídas locales posterior a tratamiento radical - Laura Bergamo y col. 201
RECAÍDAS LOCALES POSTERIOR A TRATAMIENTO RADICAL
EVALUACIÓN DE INFILTRACIÓN DEL COMPLEJO AREOLA-PEZÓN

LAURA BERGAMO, EFRÉN BOLÍVAR, ALÍ GODOY, NELLY CARRERO, LUIS BETANCOURT,
GABRIEL ROMERO, JOANNE SALAS, JUAN LEÓN
SERVICIO DE PATOLOGÍA MAMARIA. INSTITUTO DE ONCOLOGÍA “DR. LUIS RAZETTI”. CARACAS, VENEZUELA
Comunicaciones breves
201
Rev Venez Oncol 2010;22(3):201-204
Recibido: 18/11/2009 Revisado: 11/02/2010
Aceptado para publicación: 06/05/2010
RESUMEN
OBJETIVO: Experiencia de nuestra institución durante
el período 2004-2005 en recidivas locales del cáncer
de mama. MÉTODOS: Se realizó estudio clínico,
descriptivo, retrospectivo durante 2004-2005, en pacientes
con tratamiento radical y adyuvante con radioterapia,
quimioterapia si tuviesen indicación. Algunos recibieron
quimioterapia neoadyuvante. RESULTADOS: 167
pacientes sometidos a mastectomía radical. El diagnóstico
histológico correspondió 80 % (128) carcinomas ductales
infiltrantes, 4,19 % carcinomas in situ. La localización
más frecuente fue la centro-mamaria 35,48 %. El 3,65 %
fueron estadio I, 35,58 % estadio II, 53,65 % estadio III y
6,09 % estadio IV. 57 pacientes (34 %) con quimioterapia
neoadyuvante. Se realizaron 161 mastectomía radicales
tipo Madden (94,7 %), 2 mastectomía radicales tipo Patey
y 4 mastectomía total simple, 2 con ganglio centinela. 29
(17,36.%) presentaron infiltración del complejo areolapezón.
20 (68,96 %) se localizaron en región centro
mamaria. La edad promedio fue 53,6 años, 18 (62,07.%)
pacientes se ubicaron en el estadio III (37,93.%), 72,94.%
recibió quimioterapia adyuvante. El 61,76 % recibió
radioterapia, 15 (51,72 %) pacientes estaban vivas sin
enfermedad, 1 está viva con recaída local, 5 vivas con
enfermedad sistémica y 6 (20,67 %) fallecieron por
enfermedad. El seguimiento promedio fue de 36,14 meses.
Cinco pacientes del total (2,9 %) presentaron recaída
local. CONCLUSIONES: La mastectomía radical es un
arma terapéutica primaria, s índices de recaída local son
menores en centros especializados. La infiltración del
complejo aréola-pezón es más frecuente en la localización
centro-mamaria.
PALABRAS CLAVE: Cáncer, mama, mastectomía,
radical, complejo, areola pezón, recaídas, locales.
SUMMARY
OBJECTIVE: The experience in our institution during
the period 2004-2005 in local recurrence of breast cancer.
METHOD: Realized study clinical, retrospective,
descriptive during 2004-2005, in patients with radical
treatment, adjuvant radiation therapy and chemotherapy if
exist indication. Some received neoadjuvant chemotherapy.
RESULTS: 167 patients underwent radical mastectomy.
Histological diagnostic was 80 % (128) ductal infiltrante
carcinoma, 4.19 % in situ carcinoma. The more frequent
localization was mammalian center 35.48 %. 3.65 % was
stage I, 35.58 % stage II 53.65 %, stage III 6.09 % stage IV.
57 patients (34 %) with neoadjuvant chemotherapy. We
realized 161 radical mastectomy Madden type (94.7 %),
2 radical mastectomy Patey type and 4 mastectomy total
simple 2 with node sentinel. 29 (17.36 %) had infiltration
to the areola nipple zone. 20 (68.96 %) were localized in
center mammalian region. The average age was 53.6 years,
18 patients (62.07 %) were ubicated in stage III (37.93 %),
72.94 % received adjuvant chemotherapy. 61.76 % received
radiation therapy, 15 (51.72 %) patients were alive without
disease, 1 live with local recurrence, 5 staying alive with
systemic disease and 6 (20.67 %) dead by the disease. The
control average was 36.14 month. 5 patients of the total
(2.9 %) presented local recurrence. CONCLUSION: The
radical mastectomy is a therapeutically first arm, the local
recurrence index are minor in the special centers. The
infiltration of the areola nipple complex is more frequent
in the center mammalian region.
KEY WORDS: Cancer, breast, mastectomy, radical,
complex, areola, nipple, recurrence, local.
Correspondencia: Dra. Laura Bergamo.
Urb. Vista Alegre calle 2.
Edificio San Gaetano. Apto 20. Caracas, Venezuela.
Vol. 22, Nº 3, septiembre 2010
202 Recaídas locales posterior a tratamiento radical - Laura Bergamo y col.
INTRODUCCIÓN
A ctualmente el cáncer de mama
ocupa el primer lugar como causa
de muerte por cáncer en las
mujeres venezolanas (1). A pesar
del aumento de las cirugías
preservadoras, la mastectomía
radical modificada tiene aún un lugar importante
en el tratamiento primario del carcinoma de
mama. La recaída locorregional posterior a
mastectomía varía ampliamente desde 5 % a
40.% en la literatura (2). Las tasas y factores
predictivos de las recaídas locorregionales
posterior a cirugía preservadora de la mama han
sido estudiadas ampliamente, a diferencia de las
tasas y factores predictivos de recaídas posterior
a mastectomía radical modificada, las cuales han
sido estudiadas mucho menos (3).
La recaída local aislada puede ser una
fuente de metástasis a distancia o un marcador
de diseminación sistémica ocurrido antes del
tratamiento inicial (4). La influencia de varias
estrategias terapéuticas en las recaídas y su
pronóstico han sido descritos en numerosos
estudios pero, sin embargo, existe una relativa
falta de datos, de patrones de recurrencia y
pronóstico después que la recaída ocurre en
pacientes que han sido tratados con modernas
herramientas incluso tratamientos multimodales
en equipos multidisciplinarios. Sin embargo,
se ha sugerido que las mujeres que son tratadas
en centros especializados en patología mamaria
tienen mejores resultados (5).
Hay muchos estudios sobre supervivencia tras
el cáncer primario e incluso sobre la incidencia de
recidivas, pero es escasa y variable la información
disponible sobre el curso de la enfermedad y
el pronóstico después de la primera recidiva,
especialmente si la población de estudio no
queda limitada a series clínicas y se pretende
conocer esta información para el conjunto de
una población (6).
Las cifras sobre incidencia de recidivas
locorregionales varían del 9 % al 26 % a los
10 años tras mastectomía y del 7 % al 15 %
tras cirugía conservadora más radioterapia.
Las cifras de supervivencia a los 5 años tras la
recidiva locorregional se encuentran alrededor
del 35.% tras mastectomía y alrededor del 68 %
tras cirugía conservadora (7).
En el presente trabajo describimos diversos
aspectos de una serie de 2 años de pacientes con
recaídas locales posterior a mastectomía radical
en nuestra institución.
MÉTODO
Se realizó estudio clínico, descriptivo,
retrospectivo, no comparativo de pacientes
con cáncer de la mama, durante 2004-2005, en
el Instituto de Oncología “Dr. Luis Razetti”,
sometidos a tratamiento radical de la mama,
definido como la realización de mastectomía total
con márgenes negativos y los límites establecidos
en la literatura y evaluación quirúrgica de la axila
mediante la disección axilar de los niveles I y
II. El tratamiento adyuvante con radioterapia,
y quimioterapia en los casos que tuviesen
indicación. Algunos pacientes recibieron
quimioterapia neoadyuvante. Evaluando la
localización inicial del tumor y el resultado de
anatomía patológica de la pieza definitivo y la
infiltración del complejo areola-pezón.
RESULTADOS
Se evaluaron 167 pacientes sometidos a
mastectomía radical por carcinoma de la mama
en la consulta de patología mamaria del nuestra
institución entre 2004-2005. El diagnóstico
histológico correspondió en 80 % (128) con
carcinomas ductales infiltrantes, 12 % lobulillar
infiltrante, el 4,19 % fueron carcinomas ductales
in situ. La localización más frecuente de los
tumores fue la centro-mamaria con 35,48 %,
seguido del cuadrante supero-externo (13,44.%)
Rev Venez Oncol
Recaídas locales posterior a tratamiento radical - Laura Bergamo y col. 203
y unión de cuadrantes superiores (12,86 %).
El 3,65 % fueron estadio I, el 35,58 % estadio
II, el 53,65 % estadio III y el 6,09 % estadio
IV 57 pacientes (34 %) recibió quimioterapia
neoadyuvante. Se realizaron 161 mastectomía
radicales tipo Madden (94,7 %), 2 mastectomía
radicales tipo Patey y 4 mastectomía total simple,
2 con ganglio centinela. El tamaño patológico
promedio fue de 37,45 mm, el número promedio
en la disección axilar fue 14,58 ganglios.
Veintinueve (17,36 %) pacientes presentaron
infiltración del complejo areola-pezón en los
espécimen quirúrgicos. De los pacientes con
infiltración del complejo areola-pezón 20
(68,96.%) se localizaron en la región centromamaria,
y 10,34 % en unión de cuadrantes
superiores. La edad promedio de los pacientes
con infiltración areola-pezón fue 53,6 años, el
tipo histológico más frecuente en la infiltración
fue carcinoma ductal infiltrante, 18 (62,07.%)
pacientes se ubicaron en el estadio III, el
37,93.% recibió neoadyuvancia. El 72,94 %
recibió quimioterapia adyuvante. El 61,76.%
recibió radioterapia, con un estándar de
tratamiento de 5.000 cGy a la pared costal, fosas
supraclaviculares y en algunos pacientes boost
adicional a la cicatriz de la mastectomía de 1 000
cGy. 15 (51,72 %) pacientes estaban vivas libre
de enfermedad, 1 está viva con recaída local, 5
vivas con enfermedad sistémica y 6 (20,67 %)
fallecieron por enfermedad. El seguimiento
promedio fue de 36,14 meses.
Cinco pacientes del total (2,9 %) presentaron
recaída local. La edad promedio fue 61,4
años. El 80 % fueron estadio IIIA y 20 %
estadio IV. 80 % ductales infiltrantes, 60 %
presentó receptores de estrógenos positivos,
40 % receptores de progestágenos positivos,
y 60 % fueron Her2neu negativos. Solo 60.%
habían recibido radioterapia adyuvante. El
seguimiento promedio fue de 37,6 meses, una sola
paciente presentó progresión sistémica después
de la recidiva local. Todas fueron tratadas de
forma multimodal con resección local amplia,
quimioterapia y radioterapia.
DISCUSIÓN
Actualmente el cáncer de mama ocupa el
primer lugar como causa de muerte por cáncer
en las mujeres venezolanas (1). La mastectomía
radical modificada tiene aún un lugar importante
en el tratamiento primario del carcinoma de
mama. La recaída locorregional posterior a
mastectomía varía ampliamente desde 5 % a
40 % en la literatura (2). Las tasas y factores
predictivos de las recaídas locorregionales
posterior a cirugía preservadora de la mama han
sido estudiadas ampliamente, a diferencia de las
tasas y factores predictivos de recaídas posterior
a mastectomía radical modificada, las cuales han
sido estudiadas mucho menos (3).
En nuestra revisión la recidiva local estuvo
alrededor del 2 %. La recaída local aislada
puede ser una fuente de metástasis a distancia o
un marcador de diseminación sistémica ocurrido
antes del tratamiento inicial (4). La influencia de
varias estrategias terapéuticas en las recaídas y
su pronóstico han sido descritos en numerosos
estudios pero, sin embargo, existe una relativa
falta de datos, de patrones de recurrencia y
pronóstico después que la recaída ocurre en
pacientes que han sido tratados con modernas
herramientas incluso tratamientos multimodales
en equipos multidisciplinarios. Sin embargo,
se ha sugerido que las mujeres que son tratadas
en centros especializados en patología mamaria
tienen mejores resultados (5).
La mastectomía radical sigue siendo un arma
terapéutica primaria en la actualidad para el
cáncer de mama. Los índices de recaída local
son menores en centros especializados. La
infiltración del complejo areola-pezón es más
frecuente en la localización centro-mamaria,
siendo baja en otras localizaciones.
Vol. 22, Nº 3, septiembre 2010
204 Recaídas locales posterior a tratamiento radical - Laura Bergamo y col.
REFERENCIAS
1. Anuario de mortalidad. Ministerio del Poder Popular
para la Salud. República Bolivariana de Venezuela.
2006. Disponible en: URL: http//www.mpps.gob.ve.
2. Buchanan CL, Dorn PL, Fey J, Giron G, Naick
A, Mendez J, et al. Locoregional recurrence after
mastectomy. J Am Coll Surg. 2006;203:469-474.
3. Fodor J, Polgar C, Major T, Mangel LC, Szakolczai I,
Szamel I, et al. The time-course of metastases from
breast cancer after mastectomy and breast-conserving
surgery with and without isolated local-regional
recurrence. Breast. 2002;11:53-57.
4. Fodor J, Major T, Polgár C, Orosz Z, Sulyok Z, Kásler
M. Prognosis of patients with local recurrence after
mastectomy or conservative surgery for early-stage
invasive breast cancer. The Breast. 2008;17:302-308.
5. Kingsmore D, Hole D, Gillis C. Why does specialist
treatment of breast cancer improve survival? The role
of surgical management. Br J Cancer. 2004;90:1920-
1925.
6. Sarasqueta C, Martínez-Camblor P, Mendiola A,
Martínez-Pueyo I, Michelena M, Basterretxea M, et
al. Supervivencia relativa en cáncer de mama después
de la primera recidiva y factores pronóstico asociados.
Med Clin. 2009;1:1-7.
7. Clemons M, Danson S, Hamilton T, Goss P.
Locoregionally recurrent breast cancer: Incidence,
risk factors and survival [review]. Cancer Treat Rev.
2001;27:67-82.

CARCINOMA DE MAMA EN EL HOMBRE


Vol. 22, Nº 2, junio 2010
130 Carcinoma de mama en el hombre - Laura Bergamo y col.
CARCINOMA DE MAMA EN EL HOMBRE
REVISIÓN DE CASOS
LAURA BERGAMO, EFREN BOLÍVAR, ALÍ GODOY, NELLY CARRERO, LUIS BETANCOURT
SERVICIO DE PATOLOGÍA MAMARIA INSTITUTO DE ONCOLOGÍA “DR. LUIS RAZETTI” CARACAS, VENEZUELA
Comunicaciones breves
130
Rev Venez Oncol 2010;22(2):130-132
Recibido: 12/07/2009 Revisado: 02/02/2010
Aceptado para publicación: 11/03/2010
RESUMEN
El objetivo del presente trabajo es presentar el
comportamiento clínico patológico de 8 pacientes del
sexo masculino con carcinoma de la mama, atendidos en
el Instituto Oncológico “Dr. Luis Razetti” durante los años
2004-2005. El cáncer de mama en hombres representó el
2,33 %, 37,5 % fueron adenocarcinoma ductal infiltrante,
37,5 % estadio IIIA, 87,5 % se trató con mastectomías
radicales modificadas, 57,25 % expresan receptores de
estrógenos, 28,57 % sobre expresan Her2neu, 37,5 %
recibió adyuvancia, 62,5 % con tamoxifeno, el seguimiento
promedio fue de 44 meses, 37,5 % de recaídas local y
62,5.% están libres de enfermedad a los 5 años. El cáncer
de mama en hombres es muy poco frecuente. El tratamiento
primario es la cirugía, siguiendo los mismos lineamientos
para la adyuvancia que en las mujeres.
PALABRAS CLAVE: Cáncer, mama, hombre, adyuvancia,
tratamiento, cirugía, recaídas
SUMMARY
The objective of the present work is analyzing the clinic
pathologic status of 8 patients of sex male, attending in
the Oncological Institute “Dr. Luis Razetti” during the
2004-2005 years. The breast cancer in male represented
the 2.33.%, in 37.5 % of them were ductal infiltrante
adenocarcinoma, 37.5 % were state IIIA, 87.5 % were treated
with surgery: Modified radical mastectomy, 57.25 % express
estrogen receptors, 28,57 % over express Her2neu. 37.5
% received adjuvant, 62.5 % with tamoxifen, the average
of periodic control was 44 month, and 37.5 % of local
recurrence and 62.5 % of the patients are free of disease
to 5 years. The breast cancer in male is less frequent. The
primary treatment is surgery, with the same form to the
adjuvant as in women.
KEY WORDS: Cancer, breast, man, adjuvant, treatment,
surgery, recurrence.
INTRODUCCIÓN
E l carcinoma de mama masculino
es poco común ocupando 0,8 %
de todos los carcinomas de mama
(1). A pesar que puede afectar todos
los grupos etarios, el rango de
edad para el momento del
diagnóstico es entre 60 y 70 años.
El número estimado de casos nuevos y
defunciones por cáncer de mama masculino en
EE.UU en 2009 es 1 910 y 440 respectivamente(2).
En Venezuela hubo 15 defunciones en 2006 (3).
Los factores que predisponen al riesgo
parecen incluir la exposición a la radiación, la
administración de estrógenos y las enfermedades
relacionadas con el hiperestrogenismo, como la
cirrosis o el síndrome de Klinefelter (4). También
hay tendencias familiares que son definitivas,
habiéndose observado una incidencia mayor
Correspondencia: Dra. Laura Bergamo. Urb. Vista
Alegre calle 2.
Edificio San Gaetano. Apto 20. Caracas, Venezuela.
Tel. 0416725964. E-mail: laurabbrondon@hotmail.com
Rev Venez Oncol
Carcinoma de mama en el hombre - Laura Bergamo y col. 131
entre los hombres que tienen varias parientes
femeninas con cáncer de mama. Se ha informado
de un aumento en el riesgo de cáncer de mama
masculino entre las familias en las cuales se ha
identificado la mutación BRCA2 en el cromosoma
13q (5).
Clínicamente se presenta como tumor indoloro
en 75 % de los casos. El complejo areola-pezón se
encuentra afectado en 5 % a 10 % de los casos (6).
La patología es similar a la del cáncer de
mama femenino y el cáncer ductal infiltrante
es el tipo de tumor más común. También se ha
encontrado en los hombres cáncer intraductal, al
igual que carcinomas inflamatorios y enfermedad
de Paget del pezón, pero no carcinoma lobular in
situ. El compromiso de los ganglios linfáticos
y el patrón hematógeno de diseminación son
similares a los que se encuentran en el cáncer
de mama femenino (7).
El tratamiento primario estándar consiste
en una mastectomía radical modificada con
disección axilar. Las respuestas son generalmente
similares a las que se ven en las mujeres con
cáncer de mama. También se ha utilizado la
cirugía preservadora de mama (2). En los hombres
con tumores con ganglios negativos, se debe
considerar el uso de terapia adyuvante utilizando
los mismos principios que para las mujeres con
cáncer de mama, pues no hay prueba de que la
respuesta de los hombres y de las mujeres sea
diferente (8). En los hombres con tumores y
ganglios positivos, se ha utilizado quimioterapia
más tamoxifeno y otra terapia hormonal lo cual
puede aumentar la supervivencia en el mismo
grado que lo hace en las mujeres con cáncer de
mama. En la actualidad, se cuenta con estudios
controlados que comparen las opciones de
tratamiento adyuvante. Aproximadamente el
85 % de todos los tumores de mama masculino
son positivos al receptor de estrógeno y el 70.%
de los mismos son positivos al receptor de
progesterona (2,4).
Se ha utilizado terapia hormonal, quimioterapia
o una combinación de ambas con cierto éxito.
Entre estos tratamientos se encuentran la
orquidectomía, hormona liberadora de la hormona
luteinizante con bloqueo total de andrógenos o sin
este (antiandrógenos), tamoxifeno para pacientes
con receptor positivo de estrógeno, progesterona
e inhibidores de aromatasa (9).
En este estudio revisamos diversos aspectos
de una serie de 2 años de pacientes masculinos
con carcinoma de la mama en nuestra institución.
MÉTODOS
Estudio clínico, descriptivo, retrospectivo
realizado en el Instituto Oncológico “Dr. Luis
Razetti”, durante los años 2004-2005, mediante
los datos obtenidos de la revisión de historias
de los pacientes que acudieron a la consulta de
patología mamaria.
RESULTADOS
Se atendieron 342 pacientes con diagnóstico
de cáncer de mama, de los cuales 8 fueron del
sexo masculino representando el 2,33 %. La
edad promedio fue 58,12 (rango 45-80 años).
El 75.% se localizaron en la región centromamaria,
25 % en el cuadrante súpero-externo.
37,5 % fueron adenocarcinoma ductal infiltrante,
25.% carcinoma ductal in situ, 12,5 % lobulillar
infiltrante y 1 carcinoma escamoso primario de
mama. Con respecto al estadío clínico el 37,5.%
correspondió al estadio IIIA, seguido del IIB, IIIB
y 12,5 % estadio IV con metástasis pulmonar.
El 87,5 % se trató con mastectomías radicales
modificadas y el resto con cirugía preservadora, el
50 % presentó ganglios positivos en la disección
axilar niveles I, II y 1 paciente con ganglios
positivos en nivel III. El 62,5 % presentó
infiltración al complejo areola-pezón. Ninguno
presentó multifocalidad ni multicentricidad,
el tamaño patológico tumoral osciló entre
20 mm y 110 mm (promedio 48,75 mm). El
57,25 % expresan receptores de estrógenos,
Vol. 22, Nº 2, junio 2010
132 Carcinoma de mama en el hombre - Laura Bergamo y col.
el 42,85.% receptores de progestágenos, el
28,57.% sobre expresan el gen para Her2neu. El
37,5.% recibió adyuvancia tanto quimioterapia
como radioterapia, el 62,5.% tratamiento con
tamoxifeno, el seguimiento promedio fue de 44
meses, 37,5 % de recaídas local y 62,5 % están
libres de enfermedad a los 5 años, 1 paciente
falleció por progresión de enfermedad a los 46
meses de seguimiento y 1 paciente presentó
metástasis ósea.
DISCUSIÓN
En el hombre la mama es un órgano vestigial
y el cáncer de la mama en hombres es de muy
baja incidencia, creando una limitante importante
en cuanto a su etiología, evolución natural y
tratamiento. El cáncer de la mama es similar en
las mujeres que en los hombres, el diagnóstico
generalmente se realiza en estadios avanzados y
fase tardía en comparación con las mujeres. La
sobrevida global es similar a las mujeres para
cada estadio clínico-patológico.
Entre los factores de riesgo que se han
estudiado se encuentran la edad avanzada, las
alteraciones testiculares, antecedentes familiares,
exposición a radiaciones y quistes mamarios.
El carcinoma ductal invasor se ha reportado
que ocupa el 80 % de los casos, en nuestra serie
encontramos un porcentaje menor.
El tratamiento primario es la cirugía principalmente
la mastectomía radical modificada,
las conductas con respecto al tratamiento
sistémico con quimioterapia, hormonoterapia
y anticuerpos monoclonares son iguales que
para el sexo femenino. Deberían ser tratados
con radioterapia adyuvante para disminuir la
recurrencia local. El cáncer de mama en hombres
es muy poco frecuente. El tratamiento primario
es la cirugía, siguiendo los mismos lineamientos
para la adyuvancia que para las mujeres.
REFERENCIAS
1. Fentiman IS, Fourquet A, Hortobagyi GN. Male breast
cancer. Lancet. 2006;367(9510):595-604.
2. American Cancer Society. Cancer facts and figures.
Atlanta, EE.UU. 2009.
3. Anuario de mortalidad. Ministerio del Poder Popular
para la Salud. República Bolivariana de Venezuela.
2006.Disponible en: URL http/www.mpps.gov.ve
4. Giordano SH, Buzdar AU, Hortobagyi GN. Breast
cancer in men. Ann Intern Med. 2002;137(8):678-
687.
5. Wooster R, Bignell G, Lancaster J, Swift S, Seal S, Mangion
J, et al. Identification of the breast cancer susceptibility
gene BRCA2. Nature. 1995;378(6559):789-792.
6. Ribeiro GG. Carcinoma of the male breast: A review
of 200 cases. Br J Surg. 1977; 64(6):381-383.
7. Dickson RB, Pestell RG, Lippman ME. Cancer of the
breast. En: DeVita VT Jr, Hellman S, Rosenberg SA,
editores. Cancer: Principles and Practice of Oncology.
7ª edición. Filadelfia, EE.UU: Lippincott Williams &
Wilkins; 2005.p.1399-1487.
8. Golshan M, Rusby J, Dominguez F, Smith BL. Breast
conservation for male breast carcinoma. Breast.
2007;16(6):653-656.

PLAN DE TRABAJO ANTE LA AFECTACIÓN METASTÁSICA DEL GANGLIO CENTINELA EN CARCINOMA MAMARIO


Posición del Servicio de Patología Mamaria del Instituto de Oncología “Dr. Luis Razetti”

EFRÉN BOLÍVAR ABREU*, PEDRO MARTÍNEZ**, LUIS BETANCOURT*, GABRIEL ROMERO*,  ALÍ GODOY*, LAURA BERGAMO*.
SERVICIO DE PATOLOGÍA MAMARIA, INSTITUTO DE ONCOLOGÍA “DR LUIS RAZETTI”, CARACAS,VENEZUELA

*      Servicio de Patología Mamaria. IOLR
**  Jefe de Servicio de Patología Mamaria. IOLR    

Correspondencia a: Efrén Bolívar Abreu, Servicio de Patología Mamaria. Instituto de Oncología Luis Razetti. Calle Real de Cotiza, San José, Caracas. e-mail: bolivarefren@yahoo.com

RESUMEN 

   La afectación ganglionar es considerada uno de los factores pronóstico independientes más importantes en carcinoma de mama. En pacientes sin adenopatías palpables, el método de elección para determinar este estatus es la biopsia de ganglio centinela (BGC). En presencia de macrometástasis, el estándar actualmente es realizar linfadenectomía axilar (LA) a pesar de conocer que la afectación de ganglios no centinela será de un 50% aproximadamente. Cuando el compromiso ganglionar es micrometastásico, la decisión  en cuanto a realizar linfadenectomía axilar o no, se mantiene en controversia. Nos propusimos, en base a la evidencia científica existente y a nuestra experiencia, crear lineamientos que nos permitan individualizar cada caso para decidir la realización o no de linfadenectomía. Expondremos los argumentos que sustentan nuestra posición.

 Palabras clave: -Carcinoma de mama -  Ganglio centinela - Macrometástasis - Micrometástasis.

SUMMARY  

   Breast lymph node involvement  is considered one of the most important independent prognostic factors in breast carcinoma. In patients without palpable lymph nodes, the gold standard  to determine this status is the sentinel node biopsy. In the presence of macrometastases, it is now standard to perform  axillary lymphadenectomy despite knowing that the non-sentinel node involvement is approximately 50%.  When lymph node involvement  is  micrometastatic, the decision about axillary lymphadenectomy or not, remains controversial. We proposed, based on existing scientific evidence and our experience, create guidelines that allow us to individualize each case to decide whether or not performing lymphadenectomy. We will present arguments that support our position.
 Keywords: -Carcinoma of breast - Sentinel Lymph Node - Macrometastasis - Micrometastasis.




INTRODUCCIÓN

La afectación axilar es considerada como el factor pronóstico independiente más importante en cáncer de mama. En los últimos quince años se ha incrementado el número de pacientes con cáncer de mama que se han beneficiado de la BGC.1,2 Cuando éste se encuentra afectado, la linfadenectomía axilar ha sido el procedimiento de elección y esto se fundamenta en que ofrece información decisiva para el tratamiento adyuvante, funge como factor pronóstico y mejora el control local.3 La posibilidad de enfermedad adicional metastásica en la axila cuando un GC es positivo es del 40 a 50% aproximadamente. Por lo tanto, en esta situación, la práctica actual aceptada sigue siendo la LA.4 Pero por otro lado un grupo significativo de pacientes no tendrían enfermedad adicional axilar y si tenemos en cuenta morbilidad, tiempo operatorio y costos, omitir la LA sería beneficioso en estos pacientes. Por esto,  una forma de abordar este problema es relacionar los factores clínico-patológicos conocidos en cáncer de mama, con la probabilidad de enfermedad residual en la axila, con lo cual se seleccionarían los casos que no irían a LA.5
Existen otros elementos que explican el interés creciente por omitir la LA. La mayoría de estas enfermas con GC afectado recibirán quimioterapia adyuvante y su indicación no dependerá del número de ganglios con metástasis. Por otro lado, en un grupo significativo de pacientes la toma de decisiones para la planificación del tratamiento adyuvante se fundamentará en el estudio inmunohistoquímico de la tumoración, el cual posee un mayor peso en la decisión terapéutica que la propia LA. Finalmente, durante los años 2010 y 2011 han sido publicados los primeros resultados de ensayos clínicos que analizan de forma específica el impacto de la LA en el control local y la supervivencia de mujeres con GC afectado, y que no han demostrado beneficio alguno en las pacientes intervenidas con LA.6 A la luz de estos hechos, la balanza ha comenzado a inclinarse hacia una modificación en la actitud ante el GC positivo, justificando la LA sólo en casos seleccionados. 

EVIDENCIA CIENTÍFICA

El ensayo clínico multicéntrico ACOSOG Z0011 de Giuliano et al 7, muestra que las mujeres con cáncer de mama en estadio temprano y con metástasis en el ganglio centinela no se benefician de la LA. De este modo, han llegado a la conclusión de que no existen cambios en la supervivencia global ni libre de enfermedad a los 5 años, ni en la recidiva local.8 Los criterios de inclusión del estudio fueron pacientes con diagnóstico de carcinoma mamario T1/T2-N0-M0, tratadas con cirugía preservadora con 1 ó 2 GC positivos. Fueron excluídos metástasis diagnosticadas por inmunohistoquímica, pacientes con más de 2 GC positivos, plastrones ganglionares o indicación específica de radioterapia a la axila. En total fueron 891 pacientes, 445 fueron a LA y 446 no recibieron tratamiento quirúrgico adicional posterior a la BGC. El tiempo medio de seguimiento fue de 6,3 años. Todas las pacientes recibieron radioterapia en dos campos tangenciales. 
En el análisis de resultados se observó que la edad (pacientes jóvenes) y el poco grado de diferenciación, se relacionan con un mayor porcentaje de recaídas locorregionales. La afectación de ganglios no centinelas fue de un 27%, hecho observado en el brazo de pacientes sometidos a LA, lo cual hace pensar que en el grupo de pacientes de observación hubo una tendencia similar. Sin embargo, al no haber diferencias en recaídas,  resalta la importancia de los tratamientos adyuvantes como elementos de control loco regional, sugiriendo que la LA no mejora dicho control.
No obstante, hay que tomar en cuenta las limitaciones que tiene este trabajo. La principal es que la selección de pacientes fue fundamentalmente en casos con estadio muy temprano y con factores de buen pronóstico (70% con estadio T1, 83% con receptores estrogénicos positivos, 35% con micrometástasis en el GC en el brazo de LA y 44,8% con enfermedad micrometastásica en el grupo de no LA ). Además, en todos los casos se administró radioterapia a todo el lecho mamario, lo que condicionaría su posible extrapolación a campos radioterápicos menos extensos. Escaso tiempo de seguimiento, siendo la mediana de 6,3 años. El 96% de las pacientes recibió quimioterapia. No se concluyó el reclutamiento del número de pacientes previstos (891 de 1900 previstas) por razones inherentes a las mismas enfermas o negación de algunos cirujanos. El 80% de las pacientes tenían receptores hormonales positivos lo cual sesga la muestra hacia tumores de mejor pronóstico y susceptibles de manipulación hormonal. No se realizó determinación de Her2/neu ya que al inicio del estudio no era un procedimiento estándar, lo cual limita el establecimiento de subgrupos de pacientes, incluyendo a los triple negativos.6,8 
Existen otros trabajos que han estudiado la afectación del ganglio centinela. Sin embargo, la mayoría se ha dedicado al análisis de las micrometástasis. Uno de ellos es el estudio MIRROR, en el cual, casi 2600 pacientes con tumores de características biológicas favorables, sin indicación de adyuvancia sistémica con quimioterapia, con ganglios centinelas negativos pN0, células tumorales aisladas pN0(i+) o positivos para micro metástasis pN1mi, fueron separados en forma aleatoria para la realización de BGC, completar la LA o radioterapia a la axila. En este estudio omitir la disección axilar o la radioterapia a la axila en pacientes con BGC positiva para micrometástasis resultó en una mayor tasa de recaída axilar a los 5 años.9
Galimberti y col, realizaron un estudio en el Instituto Europeo de Oncología en el cual omitieron la LA en 377 pacientes con presencia de micrometástasis en GC. 2,1% (N=8) de las pacientes tuvieron recurrencias a nivel axilar. El único factor de riesgo detectado para recaída axilar fue el tamaño tumoral.10
En el ensayo NSABP-B32, dentro de los diferentes objetivos del estudio estuvo analizar los factores clínico-patológicos asociados a ganglios no centinelas positivos. El tamaño tumoral, la presencia de invasión linfovascular y la macrometástasis en el GC fueron predictores independientes. No se consideró ningún punto de corte en particular para el tamaño tumoral pues se le consideró una variable contínua (mayor tamaño, mayor afectación).11
En un estudio conducido en Barcelona, España, por Pernas y colegas, analizaron 59 casos con afectación micrometastásica del GC en las cuales no se realizó LA. No hubo recurrencias axilares en una mediana de seguimiento de 60 meses.12 En otro trabajo similar realizado por Langer y col., en 27 pacientes con presencia de micrometástasis en el GC quienes no fueron sometidas a LA, no hubo recaídas en 42 meses de seguimiento.13
Por último, en diciembre del 2011 en el  34th Annual San Antonio Breast Cancer Symposium (SABCS), la Dra. Vivianna Galimberti y col. del  International Breast Cancer Study Group (IBCSG), mostró los resultados preliminaries del IBCSG Trial 23-01, estudio llevado a cabo en 27 centros europeos en el cual 931 mujeres con presencia de micrometástasis en el GC, fueron aleatorizadas a LA o no LA. La mayoría (85%) tenía 1 ó 2 GC positivos. Con una mediana de seguimiento de 57 meses, la sobrevida libre de enfermedad fue de 87,3% y 88,4% (P= 0,48), con una sobrevida global  de 97,6% y 98% (P= 0,35) para los grupos de LA y no LA, respectivamente.17
Vemos que existen resultados variables entre los diferentes estudios, pero que en líneas generales, todos apuntan a que existen opciones seguras (Observación, radioterapia, tratamiento sistémico) que surgen como alternativas a la LA en un subgrupo de pacientes con afectación del GC. 

Experiencia del Instituto de Oncología Dr Luis Razetti
En nuestro Servicio de Patología Mamaria, nos encontramos en una búsqueda contínua de elementos de peso que sustenten nuestra práctica médica y que en definitiva se traduzcan en beneficio para los pacientes. El tema de afectación del GC ha sido uno de estos campos de investigación. En el año 2011 fue publicado un estudio por Zénzola y col.5 en el que el objetivo principal fue analizar factores clínico-patológicos en pacientes con BGC positiva, para poder predecir la afectación de ganglios no centinelas y de esta forma omitir la LA en pacientes seleccionadas. En nuestro estudio 3 factores fueron estadísticamente significativos en el análisis univariable para predecir el estado de los ganglios no centinelas: el grado nuclear III, la presencia de invasión linfovascular y el número de ganglios centinelas positivos (2 o más). Sólo la presencia de invasión linfovascular fue un factor predictor independiente (análisis multivariable) de metástasis en ganglios no centinelas.  Se concluyó  que la presencia de invasión linfovascular aumenta significativamente posibilidad de enfermedad axilar residual luego de biopsia ganglio centinela positiva. La presencia de enfermedad adicional luego de biopsia positiva es considerable (50.%). 

CONTROVERSIAS

Radioterapia
Ante la posibilidad de enfermedad residual en  la axila cuando el GC es positivo en las pacientes seleccionadas para omitir la LA, la mayoría de los centros se inclina a indicar tratamiento radiante a ese nivel como herramienta de control local. De hecho, la incidencia de recaídas axilares en mujeres con afectación del GC (micrometástasis o macrometástasis) sin LA es idéntica a las mujeres con LA cuando se asocia radioterapia postoperatoria en mama y esto se sustenta en ensayos clínicos prospectivos aleatorizados.14,15  Las pacientes que son sometidas a cirugía preservadora reciben radioterapia como parte del tratamiento integral. Los campos tangenciales inciden de forma variable en la axila dependiendo de la planificación, sobre todo a los niveles ganglionares I y II de Berg.16 Sin embargo hay que insistir al radioterapeuta en que se garantice la irradiación axilar con dosis en niveles terapéuticos en las enfermas en las que no se realice LA con el fin de mejorar el control local. En un ensayo realizado en el Instituto Europeo de Oncología, Veronesi et al., pacientes clínicamente ganglios  negativos (cN0), con tumores hasta a 1,2 cm fueron aleatorizados a recibir o no radioterapia axilar. Con una mediana de seguimiento de 63 meses, no se encontraron diferencias entre los brazos de trabajo en términos de recurrencias locales y sobrevida libre de enfermedad.18 Sin embargo, se necesita información adicional e este respecto, como la proveniente del estudio fase III  EORTC 10981-22023 After Mapping of the Axilla: Radiotherapy Or Surgery (AMAROS) trial, el cual se lleva a cabo en la actualidad, aleatorizando pacientes con GC positivo a LA o irradiación axilar. 

Uso de modelos predictivos
Se han desarrollado al menos 9 modelos matemáticos para predecir el estatus de los ganglios no centinelas en pacientes con cáncer de mama y afectación del GC. Entre ellos se incluyen 4 nomogramas (MSKCC, Mayo, Cambridge, Stanford), 3 sistemas de putuación (scoring systems) (Tenon, MD Anderson, Saidi) y 2 modelos de división recursiva (recursive partitioning) (Kohrt et al).  A través de estos métodos se intenta establecer cuantitativamente la probabilidad de metástasis en ganglios no centinelas, tomando en cuenta factores como el tamaño tumoral, tamaño de la metástasis en el GC, número de GC positivos, proporción entre el número de GC positivos y GC extirpados, invasión linfovascular, extensión extracapsular entre otros.19 El modelo que más se ha difundido en la práctica clínica y que adicionalmente ha sido validado por varios autores, es el nomograma del  Memorial Sloan-Kettering Cancer Center (MSKCC). En él se toman en cuenta 9 variables clínico-patológicas, presentando la mayor discriminación para predecir enfermedad en ganglios no centinelas.20 Estos métodos son herramientas útiles que ayudan a seleccionar pacientes para realizar o no LA. Sin embargo, muchos pierden su aplicabilidad cuando son usados fuera de la institución de origen por probables diferencias entre pacientes de diferentes latitudes y adicionalmente tienen como desventaja, el no incorporar elementos como el estatus Her2/neu, subtipos moleculares o firmas genéticas, los cuales cobran cada vez más importancia a la hora de la toma de decisiones terapéuticas en la práctica médica actual. Consideramos que pueden ser de ayuda en los diferentes grupos de trabajo, que sumado a otros factores y nunca como único elemento, sirvan como herramienta en la toma de decisiones.
Importancia de los subtipos moleculares
Desde que Perou y col, establecieron los subtipos moleculares del cáncer de mama,21 éstos han tomado cada vez mayor relevancia a la hora de establecer el pronóstico y la terapéutica de los pacientes. Las firmas genéticas, las cuales permiten identificar dichos subtipos, adicionalmente permiten establecer probabilidad de recurrencia y necesidad de tratamiento quimioterápico. Existe un alto nivel de concordancia entre los subtipos moleculares y la expresión o no de elementos fenotípicos (receptores hormonales, Her2/neu, KI 67), lo cual permite su caracterización cercana por medio de la inmunohistoquímica en los sitios en donde no se dispone de este tipo de herramientas (Oncotype, Mammaprint). 
Al establecer las características moleculares y fenotípicas del tumor, se puede instaurar un tratamiento individualizado, dirigido a ese subtipo específico, con lo cual se garantiza un mejor control de la enfermedad. Por tanto, consideramos de vital importancia su utilización en la toma de decisiones terapéuticas, tanto médicas como quirúrgicas. Un ejemplo de ello lo conforma el fenotipo triple negativo, el cual constituye un grupo de peor pronóstico con alto potencial metastásico regional y a distancia, en dónde el único tratamiento sistémico a utilizar es la quimioterapia y que por ende debe ser tratado desde el punto de vista quirúrgico de la forma más adecuada. Otros subtipos son de mejor pronóstico y susceptibles a más alternativas de tratamiento.
Resultado intraoperatorio de la biopsia de ganglio centinela
La práctica médica estándar durante la realización de la BGC es esperar el resultado de la biopsia intraoperatoria (corte congelado) y en caso que el reporte sea positivo para metástasis, proceder a la LA. Con esto se evita la posibilidad de una segunda intervención quirúrgica ya que en un solo tiempo se completa el procedimiento. Sin embargo, vemos que hay una serie de factores clínico-patológicos, algunos del tumor primario (tamaño tumoral, invasión linfovascular, etc) y otros del GC (tamaño de la metástasis, invasión extracapsular, etc) que son importantes para la toma de decisiones y que no se tienen sino posterior al estudio anatomopatológico definitivo. Esto plantea la interrogante si realizar estudio histológico peroperatorio del GC o esperar un estudio más exhaustivo para obtener la información completa y tomar la determinación más adecuada. Tanto en el MD Anderson como en el MSKCC, tienen como práctica rutinaria actual, la omisión del estudio intraoperatorio del GC, en pacientes que cumplen con los criterios del ACSOG Z0011, esperando el resultado de la biopsia definitiva para la toma de decisiones.22,23


LINEAMIENTOS PARA LA TOMA DE DECISIONES

La Sociedad Venezolana de Mastología hizo público su Consenso de Ganglio Centinela en Carcinoma Mamario en 2010. En éste se recomienda realizar LA en todos los casos de macrometástasis y deja la decisión a cargo de cada grupo de trabajo en caso de micrometástasis.  American Society of Clinical Oncology (ASCO), recomienda la realización de LA en todos los pacientes con afectación del GC, ya sean macro o micrometástasis.
 En nuestro Servicio de Patología Mamaria realizamos una discusión prequirúrgica, individualizando  la decisión para cada caso. Proponemos en base a la evidencia existente y a la experiencia propia, los siguientes criterios para omitir  la LA, en pacientes cN0 :
Tumores hasta 2 cm (T1).
Uno ó dos ganglios centinela afectados.
Ausencia de invasión linfovascular.
Garantizar la irradiación de niveles I y II de Berg.
Garantizar el tratamiento sistémico adyuvante.
Receptores hormonales positivos.
          La ausencia de tan sólo uno de los criterios indica la realización de LA.
Recomendamos esperar el resultado de  anatomía  patológica  definitivo en   los casos       en los cuales no se tengan elementos suficientes para decidir la realización o no de la LA. Sugerimos no omitir  la LA en caso de tumores triple-negativo o carcinoma Her2/neu sobreexpresado que no pueda recibir tratamiento con trastuzumab. Igualmente recomendamos la realización de la disección axilar en casos de pacientes que vayan a ser sometidas a mastectomía total (ya que la mayoría no recibirá radioterapia).

CONCLUSIONES
          Estos son los lineamientos del Servicio de Patología Mamaria del Instituto de Oncología “Dr. Luis Razetti”  para  establecer conductas  terapéuticas  en  pacientes  con  carcinoma      mamario y afectación del ganglio centinela. Los mismos se encuentran sustentados en la experiencia de nuestra institución y en evidencia científica propia y de otros centros a nivel mundial. La medicina es dinámica y los mismos irán evolucionando a medida que se vaya generando nueva evidencia. Esperamos pueda servir de guía a otros grupos de trabajo.
            BIBLIOGRAFÍA

Erb KM, Shapiro-Wright HM, Julian TB. Axillary recurrences following positive sentinel lymph node biopsy with individual tumor cells or micrometastases and no axillary dissection. Breast Dis 2010;31:83–90.

Veronesi U, Paganelli G, Viale G, Luini A, Zurrida S, Galimberti V, et al. A randomized comparison of sentinel-node biopsy with routine axillary dissection in breast cancer. N Engl J Med 2003;349:546–53.

Orr RK. The impact of prophylactic axillary node dissection on breast cancer survival: a Bayesian meta-analysis. Ann Surg Oncol. 1999;6:109–16.

Kuerer H, Newman L. Lymphatic mapping and sentinel lymph node biopsy for breast cancer: Developments and resolving controversies. J Clin Oncol. 2005;(23):1689-1705.

Zénzola V, Bolívar E, Betancourt L, Salas J, Jiménez R. Factores clínico-patológicos en cáncer de mama en relación con metástasis en ganglios no centinelas axilares cuando el centinela es positivo. Rev Venez Oncol 2011;23(3):134-144.

Acea B, et al. Propuesta de una nueva estrategia terapéutica multidisciplinar en la mujer con cáncer de mama y afectación del ganglio centinela. Cir Esp. 2011. doi:10.1016/j.ciresp.2011.09.004.

Giuliano A, Hunt KK, Ballman KV, Beitsch PD, Whitworth PW, Blumencranz PW, et al. Axillary dissection vs no axillary dissection in women with invasive breast cancer and sentinel node metastasis: a randomized clinical trial. JAMA.2011;305:569–75.

Fortea-Sanchís C. ¿Una nueva revolución en el tratamiento del cáncer de mama? Cartas al director. Cir Esp. 2012; 90(1):68-70.

Tjan-Heijnen V, Pepels M, Boer M, Borm G. Impact of omission of completion axillaries lymph node dissection or axillaries radiotherapy in breast cancer patients with micro metastasis or isolated tumor cells in the sentinel lymph node: Results from the MIRROR study. J Clin Oncol. 2009;27 (Suppl 17)(S18).

Galimberti V, Botteri E, Chifu C, Gentinline O, Luini A, Intra M, et al. Can we avoid axillary dissection in the micrometastatic sentinel node in breast cancer? Breast Cancer Res Treat. 2011. doi: 10.1007/s10549-011-1486-2.

Krag D, Anderson S, Julian T, Brown A, Harlow S, Constantino J, et al. Primary outcome results of NSABP B-32, a randomized phase III clinical trial to compare sentinel node resection to conventional axillaries dissection in clinically node negative breast cancer patients. J Clin Oncol. 2010;28:18S (Suppl) abstr LBA505).

Pernas S, Gil M, Benítez A, Bajen MT, Climent F, Pla MJ, et al. Avoiding axillary treatment in sentinel lymph node micrometastases of breast cancer: a prospective analysis of axillary or distant recurrence. Ann Surg Oncol 2010; 17:772–7.



Langer I, Marti WR, Guller U, Moch H, Harder F, Oertli D, et al. Axillary recurrence rate in breast cancer patients with negative sentinel lymph node (SLN) or SLN micrometastases: prospective analysis of 150 patients after SLN biopsy. Ann Surg 2005;241:152–8.

Fisher B, Redmond C, Poisson R. Eight-year results of a randomized clinical trial comparing total mastectomy and tumorectomy with or without irradiation in the treatment of breast cancer. N Engl J Med. 1989;320:822–8.

Giuliano A, Hunt K, Ballman K, Beitsch P, Whitworth P, Blumencranz P, et al. Axillary dissection vs no axillary dissection in women with invasive breast cancer and sentinel node metastases. JAMA. 2011;305:569–75.

Kuerer H, Newman L. Lymphatic mapping and sentinel lymph node biopsy for breast cancer: Developments and resolving controversies. J Clin Oncol. 2005;(23):1689-1705.

 Galimberti V, Cole BF, Zurrida S, Viale G, Luini A, Veronesi P, et al.International Breast Cancer Study Group Trial 23-01 Investigators. S3-1. Update of international breast cancer study group trial 23-01 to compare axillary dissection versus no axillary dissection in patients with clinically node negative breast cancer and micrometastases in the sentinel node. CTRC-AACR San Antanio breast cancer symposium, San Antanio, Texas, USA; December 6-10, 2011.

Veronesi U, Orecchia R, Zurrida S, Galimberti V, Luini A, Veronesi P, et al. Avoiding axillary dissection in breast cancer surgery: a randomized trial to assess the role of axillary radiotherapy. Ann Oncol 2005;16:383–8.


Coutant C, Olivier C, Lambaudie E, et al. Comparison of models to predict nonsentinel lymph node status in breast cancer patients with metastatic sentinel lymph nodes: a prospective multicenter study. J Clin Oncol 2009;27:2800–8.

Van Zee KJ, Manasseh DM, Bevilacqua JL, et al. A nomogram for predicting the likelihood of additional nodal metastases in breast cancer patients with a positive sentinel node biopsy. Ann Surg Oncol. 2003;10:1140–51.

Perou CM, Sorlie T, Eisen MB, et al. Molecular portraits of human breast tumours. Nature 2000;406:747-52.
Caudle A, Hunt K, Kuerer H, Meric F, Lucci A, Bedrosian I, et al. Multidisciplinary considerations in the implementation of the findings from the American college of surgeons oncology group (ACOSOG) Z0011 study: a practice-changing trial. Ann Surg Oncol. 2011;18:2407–12.

Cody III HS, Houssami N, Axillary management in breast cancer: What’s new for 2012?, The Breast (2012), doi:10.1016/j.breast.2012.01.011
















Nuevos datos del estudio de fase III EMILIA revelan que trastuzumab emtansina (T-DM1) mejoró significativamente la supervivencia en el cáncer de mama HER2-positivo metastásico



Este estudio fundamental alcanzó la variable coprincipal de valoración mejora de la supervivencia global
Roche (SIX: RO, ROG; OTCQX: RHHBY) ha comunicado hoy los resultados actualizados del estudio de fase
III EMILIA, los cuales muestran que trastuzumab emtansina (T-DM1) prolongó significativamente la vida
(supervivencia global mejorada) de mujeres con cáncer de mama (CM) HER2-positivo metastásico en
comparación con la combinación de lapatinib y Xeloda (capecitabina). El estudio EMILIA, en pacientes con
CM HER2-positivo metastásico tratadas previamente con Herceptin (trastuzumab) y un taxano
(quimioterapia), ha alcanzado ahora ambas variables principales de eficacia: mejorar tanto la supervivencia
global (SG) como la supervivencia sin progresión (SSP). Estos datos se presentarán en un próximo congreso médico.
Genentech ha solicitado a la FDA –el organismo regulador de los medicamentos en los Estados Unidos– una
autorización de producto biológico para trastuzumab emtansina y Roche solicitará en breve a la EMA –
Agencia de Medicamentos de la Unión Europea– la correspondiente autorización de comercialización.
«Nos complace extraordinariamente comunicar que mujeres tratadas con trastuzumab emtansina vivieron
significativamente más tiempo que las que recibieron un tratamiento estándar contra este agresivo cáncer de
mama avanzado –ha comentado Hal Barron, director médico y director de Desarrollo Internacional de
Roche–. Estamos convencidos de que los conjugados anticuerpo-fármaco tienen el potencial para cambiar el tratamiento del cáncer en el futuro, y nos alegramos de colaborar con las autoridades sanitarias para ofrecer otra opción terapéutica potencial a las mujeres con cáncer de mama HER2-positivo metastásico».
Considerando los resultados globales actualizados sobre la supervivencia del estudio EMILIA, a las pacientes del grupo lapatinib + Xeloda se les ofrecerá la posibilidad de recibir trastuzumab emtansina.
Trastuzumab emtansina es un conjugado anticuerpo-fármaco (ADC por sus siglas en inglés) en investigación
contra cánceres HER2-positivos. Trastuzumab es el anticuerpo y DM1 el quimioterápico, unidos por un
acoplador estable. Trastuzumab emtansina actúa inhibiendo selectivamente la vía de señalización de HER2 y
llevando el agente quimioterápico DM1 directamente al interior de las células cancerosas HER2-positivas.
Desde hace tres decenios estudia Roche la vía de señalización de HER2. El desarrollo de terapias de acción
selectiva sobre HER2 constituye uno de los primeros ejemplos de los beneficios de la medicina personalizada.
Acerca del estudio EMILIA
EMILIA (TDM4370g/BO21977) es un estudio de fase III abierto, aleatorizado e internacional en el que se
compara trastuzumab emtansina en monoterapia con la combinación de lapatinib y Xeloda en 991 mujeres
con cáncer de mama HER2-positivo localmente avanzado o metastásico que habían recibido previamente
Herceptin y quimioterapia basada en taxanos.
Las variables principales de eficacia del estudio son la SSP (evaluada por un comité de investigación clínica
independiente) y la SG. Otras variables de valoración del estudio son la tasa de supervivencia al cabo de uno y dos años, el perfil de seguridad, la SSP evaluada por los investigadores, la tasa de respuesta objetiva, la duración de la respuesta y la calidad de vida.
Resultados globales actualizados sobre la supervivencia en el estudio EMILIA
Los resultados del análisis confirmatorio de la SG en el estudio de fase III EMILIA excedían el límite
preespecificado, demostrando que trastuzumab emtansina alargaba significativamente la vida de las mujeres
con CM HER2-positivo metastásico en comparación con la combinación de lapatinib y Xeloda.
Resultados del estudio EMILIA presentados con anterioridad
Resultados del estudio EMILIA se presentaron en junio de 2012 en el 48.º congreso anual de la Sociedad
Americana de Oncología Clínica (ASCO)1:
• De acuerdo con la evaluación de un comité independiente, en las pacientes tratadas con trastuzumab
emtansina (n = 495) hubo una mejora significativa del tiempo que vivieron sin empeorar (SSP) en
comparación con las que recibieron lapatinib + Xeloda (n = 496).
• El riesgo de empeoramiento de la enfermedad o de muerte disminuyó en un 35% en las pacientes tratadas
con trastuzumab emtansina en comparación con las que recibieron lapatinib + Xeloda (hazard ratio [HR]:
0,65, p <0 6="6" 9="9" a="a" de="de" frente="frente" mediana="mediana" meses="meses" p="p" respectivamente="respectivamente" ssp:="ssp:">• Pacientes con acontecimientos adversos (AA) de grado 3 o superior (graves) hubo menos entre las
tratadas con trastuzumab emtansina que entre las que recibieron lapatinib + Xeloda: 40,8% y 57,0%,
respectivamente. En comparación con las pacientes tratadas con lapatinib + Xeloda, los AA de grado 3 o
superior más frecuentes (observados en más del 2% de las pacientes) en las que recibieron trastuzumab
emtansina consistieron en recuento bajo de plaquetas (12,9% frente al 0,2%), aumento de las cifras de
enzimas liberadas por el hígado y otros órganos (aspartato-aminotransferasa: 4,3% frente al 0,8%; alaninaaminotransferasa:
2,9% frente al 1,4%) y anemia (2,7% frente al 1,6%).
• En un análisis intermedio previo de la supervivencia global quedó demostrada una tendencia hacia una
SG mejor en las mujeres tratadas con trastuzumab emtansina que en las tratadas con lapatinib + Xeloda.
Ahora bien, los datos no se consideraron estadísticamente significativos en ese momento.
Acerca de trastuzumab emtansina Trastuzumab emtansina es un conjugado anticuerpo-fármaco (ADC) en investigación contra cánceres HER2-positivos. Trastuzumab es el anticuerpo y DM1 el agente quimioterápico, unidos por un acoplador estable. Trastuzumab emtansina tiene los mecanismos de acción de ambos: trastuzumab y DM1.
Trastuzumab emtansina actúa inhibiendo selectivamente la vía de señalización de HER2 y llevando el agente
quimioterápico DM1 directamente al interior de las células cancerosas HER2-positivas. Trastuzumab se une
a las células cancerosas HER2-positivas y se cree que bloquea las señales incontroladas favorecedoras del
crecimiento del cáncer, estimulando a la vez el sistema inmunitario del organismo para atacar las células
cancerosas. Trastuzumab emtansina está diseñado para que, una vez absorbido hacia el interior de las células, las destruya mediante la liberación del DM1.
Genentech, miembro del Grupo Roche, ha adquirido en licencia la tecnología de trastuzumab emtansina en
un acuerdo con ImmunoGen, Inc.
Roche tiene en su cartera de desarrollo unos 25 conjugados anticuerpo-fármaco, en los que aprovechará la
experiencia adquirida en los estudios con trastuzumab emtansina.
Acerca del cáncer de mama El cáncer de mama es la neoplasia maligna más frecuente en las mujeres en todo el mundo1. Cada año se diagnostican aproximadamente 1,4 millones de nuevos casos de cáncer de mama y mueren más de 450.000 mujeres de esta enfermedad2. Este tipo de cáncer se caracteriza por la presencia de cantidades elevadas de HER2 (receptor 2 del factor de crecimiento epidérmico humano) en la superficie de las células tumorales. Se habla entonces de «positividad para HER2», la cual se detecta en el 15-25% de las mujeres con cáncer de mama3. El cáncer HER2-positivo es una forma particularmente agresiva de cáncer de mama4.

Acerca de Roche
Roche, cuya sede central se halla en Basilea (Suiza), es una compañía líder del sector de la salud, centrada en la investigación y con la potencia combinada de la integración farmacéutica-diagnóstica. Roche es la mayor empresa biotecnológica del mundo y tiene medicamentos auténticamente diferenciados en las áreas de oncología, virología, inflamación, metabolismo y sistema nervioso central. Roche también es el líder mundial en diagnóstico in vitro, incluido el diagnóstico histológico del cáncer, y pionero en el control de la diabetes.

Referencias
1. Blackwell K et al Primary results from EMILIA, a phase 3 study of trastuzumab emtansine (T-DM1) vs capecitabine (X) and
lapatinib (L) in HER2-positive locally advanced or metastatic breast cancer (MBC) previously treated with trastuzumab (T) and
a taxane. ASCO 2012 Abstract LBA1.
2. Ferlay J, et al GLOBOCAN 2008, Cancer Incidence and Mortality Worldwide: IARC Cancer Base No. 10 [Internet]. Lyon,
France: International Agency for Research on Cancer; 2010. Available from: http://globocan.iarc.fr.
3. Wolff A.C et al. American Society of Clinical Oncology/College of American Pathologists Guideline Recommendations for
Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer. Arch Pathol Lab Med 2007: 131.
4. Slamon D et al. Adjuvant Trastuzumab in HER2-Positive Breast Cancer. N Engl J Med 2011; 365:1273-83.

La radiación diagnóstica torácica podría aumentar el riesgo de cáncer de mama


EN BRCA1 Ó BRCA2 POSITIVAS


Las mujeres portadoras de mutaciones en BRCA1 o BRCA2 que se han sometido a radiación torácica diagnóstica (mamografía, radiografía, TC...) antes de los 30 años tienen mayor riesgo de cáncer de mama que las que presentan esas mismas mutaciones genéticas pero no han estado expuestas a radiación. Así lo revela un estudio que se publica en British Medical Journal (BMJ).

Los autores del trabajo, dirigido por Flora van Leeuwen, del Instituto del Cáncer de Holanda, señalan que algunos estudios han sugerido que las mujeres con mutaciones en BRCA1/2 podrían tener mayor sensibilidad a la radiación porque estos genes están implicados en la reparación del daño en el ADN, que puede estar causado, precisamente, por la radiación.
Por lo tanto, los beneficios del cribado mamográfico en las portadoras más jóvenes serían menores que los riesgos.
El estudio que recoge BMJ examinó la cuestión en 1.993 mujeres. Todas ellas eran portadoras de alguna de las citadas alteraciones genéticas que confiere un mayor riesgo de cáncer de mama. El seguimiento se dio por finalizado con el primer diagnóstico de cáncer de mama.
Al 43 por ciento de ellas se les diagnosticó un cáncer de mama, el 48 por ciento se había sometido alguna vez a una radiografía y el 33 por ciento a una mamografía. La historia de exposición a radiación torácica entre los 20 y los 29 años incrementaba el riesgo de cáncer de mama en un 43 por ciento, cifra que ascendía al 62 por ciento cuando se había iniciado antes de los 20. Los autores señalan que sus resultados deben ser validados por estudios más amplios.
(British Medical Journal. DOI: 10.1136/bmj. e5660).

¿POR QUÉ NO HAN DESCUBIERTO LA CURA CONTRA EL CÁNCER?


¿POR QUÉ NO HAN DESCUBIERTO LA CURA CONTRA EL CÁNCER?
Probablemente en la actualidad no exista una respuesta tangible a esta pregunta, pero contestando de manera realista, yo diría que algunos tipos de cáncer sí son curables si se detectan a tiempo. En otros casos ni siquiera sabemos exactamente cómo se desarrollan. Lo que sí es seguro es que cada vez lo conocemos mejor y lo derrotamos en más batallas. Esperamos algún día ganarle la guerra.
La investigación en oncología avanza muy rápido. Hace poco menos de 20 años no sabíamos muchas cosas que ahora manejamos, y la herramienta fundamental para llegar a estos conocimientos ha sido una disciplina definida como genómica, la cual estudia todos los genes de un ser vivo. Ésta, de la mano de la proteómica y la biología molecular, nos ha permitido entender una serie de fenómenos que explican el origen, desarrollo e interacciones del cáncer. El descubrimiento de nuevos genes, proteínas y vías de señalización, forman parte de la gran cantidad de información de la cual se dispone en la actualidad. Incluso podemos calcular la posibilidad que tiene una persona de desarrollar cierto tipo de tumor en base a su carga genética.
cura-cancerMúltiples fármacos han sido creados para actuar en receptores y complejos proteicos (denominados dianas terapéuticas) y de esta forma interferir en el desarrollo de células cancerígenas. Además, las campañas de pesquisa han permitido detectar algunos tipos de neoplasias malignas en etapas tempranas de su historia natural, con lo cual logramos curar a una cantidad importante de pacientes. Entonces sí es real aquel emblemático eslogan que dice que "el cáncer es curable si se diagnostica a tiempo". Pero, ¿por qué no se ha descubierto una cura global contra todos los tipos de cáncer o una vacuna que ni siquiera permita que se desarrolle alguno de ellos?
No todos los cánceres son iguales. Existen unos más agresivos que otros y con sensibilidades diferentes a los tratamientos. Por otro lado, no todos los seres humanos se comportan de la misma manera ante esta enfermedad. Esto podría servir de asidero para generar una respuesta hipotética a nuestra pregunta principal: no curamos el cáncer de forma global; cuando lo hacemos, lo logramos de forma individualizada.
Mientras tanto, seguimos encarnando una lucha constante contra este mal que aqueja a millones de personas en el planeta, una enfermedad heterogénea y difícil de descifrar, cuyo mayor entendimiento nos impulsa a seguir transitando un camino, que aunque sabemos será largo y tortuoso, nos llevará a la meta tan anhelada: curar definitivamente el cáncer.


Efrén Bolívar Abreu

Efrén Bolívar Abreu

MD. Cirujano Oncólogo. Mastólogo. PhD en Ciencias Médicas. Máster en Patología Mamaria Universidad de Barcelona - España. Profesor de Técnica Quirúrgica de la Universidad Central de Venezuela. En Twitter @efrenbolivar.

Current issues in diagnostic breast pathology


Rosemary A Walker,1 Andy Hanby,2 Sarah E Pinder,3 Jeremy Thomas,4
Ian O Ellis,5 National Coordinating Committee for Breast Pathology Research
Subgroup, On behalf of members of the National Coordinating Committee for
Breast Pathology
ABSTRACT
On behalf of the NHS Breast Screening Programme
Pathology Coordinating Group we present
recommendations for terminology and diagnostic criteria
for a number of key areas of practice in breast pathology
where terminology can be confusing and where accurate
communication will ensure appropriate clinical
management. These recommendations cover columnar
cell lesions and the spectrum of changes that can be
seen in these epithelial proliferations, lobular neoplasia,
micrometastases and isolated tumour cells in axillary
lymph nodes, the use of basal/myoepithelial markers in
diagnostic practice and oestrogen receptor testing in
ductal carcinoma in situ.
INTRODUCTION
The research subgroup of the NHS Breast Screening
Programme (NHSBSP) Pathology Coordinating
Group have compiled overviews and recommendations
for topics that were either not considered or
had minimal discussion in NHSBSP publication
number 58 Pathology Reporting of Breast Disease.
These will be included as appendices in the pending
revision of the non-operative reporting guidelines.
In conjunction with NEQAS the HER2 reporting
guidelines have been updated and published:Walker
RA, Bartlett JM, Dowsett M, et al. HER2 testing in
the UK: further update to recommendations. J Clin
Pathol 2008;61:818e24.
These topics are emerging issues in diagnostic
breast pathology and are presented below as a series
of short monographs.
COLUMNAR CELL LESIONS: NHSBSP
COORDINATING GROUP RECOMMENDATIONS FOR
TERMINOLOGY AND DIAGNOSTIC CRITERIA
Background
In recent years there has been growing interest and
awareness of epithelium within breast biopsies
having a columnar morphology. One of the common
accompaniments of this change is the presence
of secretions within lumina, which are often
distinctly eosinophilic and associated with microcalcifications.
1 These microcalcifications are detectable
at mammography and consequently more of
these lesions are being detected with the introduction
and expansion of breast screening programmes.
The diverse terminology employed for this
epithelium, its increased detection at screening and
the uncertainties regarding its biology continue to
cause concern among both diagnostic breast
pathologists and their clinical colleagues in the
breast care team. In this document, we present
a proposal for a simplified terminology for lesions
with columnar epithelium and histological criteria
for recognising these, and recommendations for
classification using the ‘B’ grading system in core
biopsies. These proposals are not set in stone and
may change as more knowledge is acquired
regarding the pathobiology of these lesions and
their relationship to malignancy.
Terminology
It should be noted that a columnar/flat morphology
is often seen in the epithelium of benign breast
lesions outside the columnar change/flat epithelial
atypia (FEA) spectrum and in cytological preparations
from a number of conditions, for example,
intraductal papillomas.2 Tall columnar cells are also
a feature of a number of more unusual carcinoma
variants, for example, a tumour recently described
as resembling the tall cell variant of papillary
thyroid carcinoma.3
Here we refer to a group of lesions deduced by
several authors to lie within the constellation of
fibrocystic change.4 5 Within this group, a more
focused spectrum of lesions has been delineatedd
namely, columnar cell change (CCC), CCC with
hyperplasia, CCC with atypia and columnar cell
hyperplasia with atypia.6 The morphology of those
with atypia corresponds to FEA, as detailed in the
latest WHO classification.7
CCC5 8e10 has been otherwise variously described
and/or illustrated over the years and lesions with
attributes of columnar lesion have been referred to
under a great diversity of names.1 4 7 11e23 Some of
these terms are detailed in figure 1. However, in
assessing these it is worth noting, as Ho et al
comment, that ‘one must be extremely careful
when comparing outcome of FEA and its equivalents
in the literature, without first having determined
if authors are referring to the same lesion’.23
This statement equally applies to all forms of the
overlapping columnar cell lesions (CCLs).
The proposed ductal intraepithelial neoplasia
(DIN) spectrum described by Bratthauer et al
ranges from usual type ductal hyperplasia through
to high-grade ductal carcinoma in situ (DCIS),24
although molecular studies do not support this as
a continuum and this system of classification is not
widely used. The territory mapped out relevant to
the present discussion is the DIN 1-flat type which
equates to FEA.24
More recently, terminological practice has been
generally simplified (figure 2), reflecting current
guidelines.
1Cancer Studies and Molecular
Medicine, University of
Leicester, Leicester, UK
2Academic Unit of Pathology,
Leeds University, Leeds, UK
3Academic Oncology/Breast
Pathology, King’s College
London, London, UK
4Department of Pathology,
Western General Hospital,
Edinburgh
5Molecular Medical Sciences,
University of Nottingham,
Nottingham, UK
Correspondence to
Dr Ian O Ellis, Molecular Medical
Sciences, University of
Nottingham, Department of
Histopathology, Nottingham City
Hospital, Hucknall Road,
Nottingham NG7 1DD, UK;
ian.ellis@nottingham.ac.uk
Accepted 1 February 2012
J Clin Pathol 2012;65:771–785. doi:10.1136/jclinpath-2012-200733 771
Review
Published Online First
19 July 2012
Downloaded from jcp.bmj.com on September 17, 2012 - Published by group.bmj.com
Microscopic appearances of CCLs (Table 1)
CCCs without atypia
The classic CCL is comprised of lobular acini lined by epithelial
cells that are tall and snouted in a manner similar to that
observed in tubular carcinoma. Commonly this is associated
with luminal secretions and/or microcalcifications (figure 3A,B).
If there is a piling up of several layers the term ‘columnar cell
hyperplasia’ is used, assuming that the stratification is real
(figure 3D) rather than artefactual as a consequence of crosscutting
(figure 3C). There is morphological diversity within
these groups, for example, the hyperchromasia of the nuclei can
vary, as well as nuclear shape and the ‘tallness’ of the cells; thus
in some cases some lesions are more cuboidal than columnar.
Flat epithelial atypia
If cytological atypia is present in CCC or columnar cell hyperplasia,
the lesions should be classified as FEA (figure 2).8 33 There
is, however, morphological diversity in this group of lesion; in
some cases the atypia manifests as relatively uniform rounded
evenly spaced nuclei with a similar cytomorphology to that
displayed by the cells of low and some intermediate grade DCIS
(figure 4C). In this form of FEA, polarisation of the cells may be
lost. Nuclei are often more centrally placed. In others, distinct
nuclear pleomorphism is evident, falling short of high nuclear
grade but, equally, without the relative nuclear uniformity
described above (figure 4A,B). Finally, some FEA display
a distinctly endometrioid appearance with tightly packed tall
hyperchromatic fusiform nuclei (figure 4D).
Exclusions
Atypical ductal hyperplasia
FEA by definition does not have a complex architecture; if
micropapillary or cribriform structures are seen, the diagnosis of
atypical ductal hyperplasia (ADH) or low-grade DCIS should be
considered.34 The process may involve more than one terminal
duct lobular unit. Sporadic atypical architecture may be widely
spaced. Rather than result in overdiagnosis of DCIS, it is proposed
that the current size criterion guidelines for differentiating DCIS
and ADH (>2 mm or >2 duct spaces)25 be relaxed and the term
FEA be applied under these circumstances. Whether most FEA is
biologically equivalent to most ADH is, as yet, uncertain but
likely. In practice, FEA is often seen merging with the more
elaborate architecture of ADH (figure 5) and DCIS, demonstrating
the close relationship of FEA to these lesions. Lesions which fulfil
the criteria for low-grade DCIS by showing $2 ducts fully
involved by an architecturally and cytologically atypical process
without intervening gaps of non-atypical epithelium should still
be regarded as such and not as FEA.
Ductal carcinoma in situ
Where flat epithelial proliferative epithelium within a terminal
ductal lobular unit (TDLU) possesses high cytonuclear grade
nuclei, the lesion should be categorised as high-grade flat DCIS,
not FEA (figure 6). Care should be taken in the diagnosis of flat
high-grade DCIS in core biopsies and it may be prudent to
categorise this as B4 suspicious in such limited samples.
Differential diagnosis
It should be borne in mind that there are many phenomena with
low or ‘flat epithelium’ that do not fall under the category of the
CCLsdfor example, the tall monolayered epithelium of in situ
papillary neoplasia.
Apocrine change
Apocrine epithelium shares with columnar change the presence
of apical snouts and sometimes associated secretions. However,
the cytoplasm of apocrine cells is typically copious and granular
and, unlike most columnar cells, nucleoli are readily seen.
Apocrine cells are usually androgen receptor positive but
oestrogen receptor (ER) and progesterone receptor (PR) negative;
the converse is true for columnar epithelium.
Lactational change
Another lesion that may be mistaken for FEA is focal lactational
change, which may appear both monolayered and mildly
cytologically atypical with enlarged hyperchromatic nuclei.
Typically, the epithelial cells are more cuboidal than tall and
the foamy appearance to the cytoplasm produced by finely
divided lipid should alert the pathologist to this diagnosis
(figure 7).
Markers in CCLs
Producing a clear idea of the marker profile in columnar lesions is
bedeviled by the variety of terminology used in different
Figure 1 Varied terminology-related lesions with columnar and flat
epithelium. ADH, atypical ductal hyperplasia; DCIS, ductal carcinoma in
situ; DIN, ductal intraepithelial neoplasia.
Figure 2 Most simple classification of columnar lesions (after Schnitt
and Vincent-Salomon6). DCIS, ductal carcinoma in situ.
772 J Clin Pathol 2012;65:771–785. doi:10.1136/jclinpath-2012-200733
Review
Downloaded from jcp.bmj.com on September 17, 2012 - Published by group.bmj.com
publications and the uncertainty about their equivalence. What
follows is a distillation of what is known, under the broad
umbrella of the names indicated above.
Unlike the normal breast and usual ductal hyperplasia (UDH)
where ER and PR staining is heterogeneous and limited to about
10e15% of cells,25 in CCC and related lesions there is extensive
and uniform positivity of the nuclei of all lesional cells.4 5 15 20
Most cells stain positively for keratin 1915 and osteoprotegerin26
and a high proportion stain for cyclin D1.15 Kusama et al
demonstrated a Ki67 labelling index of 0.3% in columnar lesions
versus 13.7% in adjacent DCIS.18 Unlike UDH, the lesional cells
do not express basal markers such as CK5/6.27 As indicated
above, immunohistochemistry (IHC) may be useful in separating
apocrine from columnar epithelium, the former being
Figure 3 Columnar cell change
without atypia (A-D) Columnar cell
change affects lobular units which show
variably dilated acini often with
microcystic change and calcification
(figure 3A, B). The lining epithelial
morphology varies from low cuboidal to
the classic columnar type with apical
snouts (3C) In more florid cases the
epithelium is multi-layered and
classified as columnar cell hyperplasia
without atypia (3D).
Table 1 Comparison of pathological features of the spectrum of entities from columnar cell change through to ADH/DCIS
Feature
Diagnosis
Columnar cell change Columnar cell hyperplasia FEA ADH/DCIS
Topography TDLU, acini may be mildly
dilated or of normal size
TDLU, acini may be mildly
dilated or of normal size
TDLU, often microcystically
dilated acini
TDLU 6 adjacent ducts
Shape of acinar spaces Irregularly shaped luminal margin Irregularly shaped luminal margin Often rounded acinar spaces,
with smooth inner margin
Often rounded acini, but with
complex structures extending
into lumen (see architecture,
below).
Architecture Flat Tufts and mounds Flat or tufted, not complex Complex with micropapillary
or cribriform structures
Stratification/multilayering Not present Present May be present May be present
Luminal secretions often
with microcalcification
Present Present Present May be present
Nuclear size Small to medium Small to medium Small to medium Small to medium
Nuclear shape Oval, elongated Oval, elongated Often, but not always, rounded Rounded
Nuclear texture Bland Bland Speckled chromatin pattern
may be present
Speckled chromatin
pattern common
Pleomorphism* Uniform Uniform Uniform to moderately
pleomorphic
Uniform
Position of nuclei within cell Basally placed Basally placed Often central Central
Nucleoli Not conspicuous Not conspicuous Not conspicuous, but may
be evident
Not conspicuous, but may
be evident
Mitoses Generally absent Generally absent Generally scarce Generally scarce
Extent May be focal or extensive May be focal or extensive May be a focal area
within background of
non-atypical CCL
May be focal area within
background of non-atypical
CCL. By definition ADH is
small/microfocal
*If marked pleomorphism, the lesion does not fall with the spectrum of CCLs but should be regarded as high-grade DCIS.
ADH, atypical ductal hyperplasia; CCLs, columnar cell lesions; DCIS, ductal carcinoma in situ; FEA, flat epithelial atypia; TDLU, terminal ductal lobular unit.
J Clin Pathol 2012;65:771–785. doi:10.1136/jclinpath-2012-200733 773
Review
Downloaded from jcp.bmj.com on September 17, 2012 - Published by group.bmj.com
androgen receptor positive and ER and PR negative. The utility
of GCF15, typically positive in apocrine epithelium, in making
this distinction has yet to be documented in the literature. It is
worth noting that McLaren et al found that patients with ER
negative CCLs were at increased risk of malignancy compared
with those whose lesions were ER positive,23 but as yet there are
no confirmatory studies.
In conclusion, at present the main use for IHC is in the
differential diagnosis of columnar from some apocrine lesions.
Significance of CCLs
CCLs of the breast encompass a morphological spectrum which
may coexist within a wide range of lesions that may include in
situ and invasive carcinoma.9 However, the overall biological
significance of these lesions is unknown.28 There is supporting
evidence for a precursor role including an association with
tubular carcinoma9 12; the finding that these lesions are found
more commonly in cancerous than non-cancerous breasts17; are
often continuous with cancerous lesions18; commonly coexist
with lobular in situ neoplasia22; and share similar cytological
characteristics and immunohistochemical profile with coexistent
malignancies.15
Figure 5 Atypical ductal hyperplasia. Flat epithelial hyperplasia,
particularly when florid can show both cytonuclear and architectural
feature of low grade DCIS but fail to have sufficcent extend for
classification as DCIS. The diagnosis of atypical ductal hyperplasia
arising in a background of FEA can be made in these circumstances.
Figure 6 High-grade ductal carcinoma in situ may show mono- or
oligo-layering within the inner surface of an involved duct. High-grade
nuclear pleomorphism such as this is not seen in flat epithelial atypia.
Figure 4 Flat epithelial atypia (4 A-D)
Flat epithelial atypia usually shows
more exajurated involvement of the
breast parenchyma (4A). The cells
show a loss of nuclear polarity but may
retain a columnar morphology (4B). The
nuclei are often more rounded and less
ovoid than in columnar cell change
(4C0. The degree of atypia can become
more marked but is not high grade (4D)
774 J Clin Pathol 2012;65:771–785. doi:10.1136/jclinpath-2012-200733
Review
Downloaded from jcp.bmj.com on September 17, 2012 - Published by group.bmj.com
Despite the above, the certainty and speed of progression of
these lesions to malignancy is unknown. Eusebi et al11 discovered
25 cases (0.26%) of what was called low-grade clinging
carcinoma of flat type, but which appears morphologically
equivalent to FEA. During an average follow-up interval of
19 years, none of the 25 women with low-grade clinging carcinoma
of flat type developed invasive carcinoma and only one
had a recurrence, which was described as consisting of low-grade
clinging carcinoma of flat type.29
At a more fundamental level, there is evidence that, in general,
there is a progressive accumulation of allelic damage in FEAthrough
to DCIS and invasive carcinoma, implying a molecular continuum
from at least some lesions with a columnar morphology (which is
not the case for UDH and carcinoma).28 30 Despite the very limited
follow-up data to date and the need for more evidence, this implies
that the biological progression of an individual columnar lesion
may be slow. However, it seems very likely that FEA is one of the
earliest forms of neoplastic change in the breast.
CCLs in breast core biopsies
Cores bearing CCLs are typically sampled for the histological
assessment of mammographic microcalcifications. As for other
such specimens, these should be examined at multiple levels (at
least three). If CCC or hyperplasia only is found without atypia,
the lesions should be regarded as within the constellation of
fibrocystic change and categorised as B2, benign.
CCLs with atypia should be regarded as FEA and classified as
B3, of uncertain malignant potential. Lesions with more
complex architecture should also be regarded as an atypical
epithelial proliferation and also regarded as B3, of uncertain
malignant potential.
As for all such screen-detected lesions, multidisciplinary
discussion should be undertaken to correlate radiological, clinical
and histopathological findings. Data on risk of finding adjacent
associated malignancy are extremely limited.
CCLs in surgical excision specimens
Thorough sampling and histological examination of surgical
specimens bearing FEA should be performed to search for more
established neoplasia. In association with some DCIS and/or
invasive carcinoma, FEA may be present and may be extensive
and/or may extend to the margins of the specimen. This is
particularly problematic for lesions which are well recognised as
being associated with FEA such as tubular carcinoma9 and, in
such cases, it may be uncertain as to whether the CCL represents
the precursor process. There is a very limited evidence base
for guidance but, at present, it is recommended that whole
tumour size (DCIS plus invasive carcinoma) and margin status
should include only those areas regarded as established DCIS or
invasive tumour using conventional criteria.
Reproducibility of current classification of CCLs
The reproducibility of diagnosis of CCL varies in the published
literature. One study looking at the assessment of images by
a mixed population of assessors showed poor agreement.31
Another demonstrated ‘excellent agreement’ for a specified task
within the CCL group after a PowerPoint directed training
session undertaken by a group of pathologists with an interest in
breast pathology.32 Agreement was slightly better for determining
the absence of FEA (92.8%, 95% CI 84.1% to 97.4%) than
for determining its presence (90.4%, 95% CI 79.9% to 96.7%).
The latter authors concluded that the diagnosis of FEA and its
distinction from CCLs without atypia was highly reproducible if
the presently available diagnostic criteria were adhered to. This
simplified classification in the latter study forms the basis of the
recommendations presented here.
Issues of poor reproducibility can confound diagnosis and lead
to inconsistent patient care. This is more likely when complex
and diverse terminology with shades of possible interpretations
exist. This is no more apparent than in the case of lesions
possessing columnar epithelial morphology and is the strongest
argument for the simplified classification of these lesions
presented in this document.
IN SITU LOBULAR NEOPLASIA: NHSBSP COORDINATING
GROUP OVERVIEW AND RECOMMENDATIONS
Lobular carcinoma in situ (LCIS) was first described in 1941 by
Foote and Stewart33 and was considered to be akin to DCIS with
Key points and recommendations
< The terms columnar cell change and columnar cell hyperplasia
are regarded as benign lesions falling within the constellation
of fibrocystic changes and in core biopsy should be classified
as B2, benign.
< Columnar cell lesions with cytological atypia should be
regarded as FEA and, in a core biopsy, categorised as B3, of
uncertain malignant potential.
< Sporadic atypical architecture may be widely spaced. Rather
than result in overdiagnosis of DCIS, it is proposed for the
current guidelines that the size criterion for differentiating
DCIS and ADH (>2 mm or >2 duct spaces)25 be relaxed and
the term FEA be applied under these circumstances. In needle
core biopsy samples these should be regarded as B3, of
uncertain malignant potential.
< Columnar cell lesions with high cytonuclear grade should be
regarded as flat high-grade DCIS.
Figure 7 (A) Lactational changes can
be mistaken for flat epithelial atypia and
(B) in some rare variants show distinct
cytological atypia. The epithelium tends
to be cuboidal and the presence of
foamy clear cytoplasm should alert one
to this diagnosis.
J Clin Pathol 2012;65:771–785. doi:10.1136/jclinpath-2012-200733 775
Review
Downloaded from jcp.bmj.com on September 17, 2012 - Published by group.bmj.com
regard to clinical behaviour, with the recommended management
being mastectomy. Subsequently, lobular neoplasia, the
term introduced by Haagensen et al,34 was felt to differ from
DCIS, being a risk indicator of subsequent development of breast
cancer34e36 rather than a true precursor, so radical surgical
treatment fell out of favour. The advent of mammographic
screening, further follow-up studies and molecular analysis of
lobular neoplasia has resulted in the need to re-address its
significance and management.
Terminology
Atypical lobular hyperplasia (ALH) and LCIS have been considered
distinct entities. They are both characterised by a proliferation
within the TDLUs of poorly cohesive, monotonous
cuboidal or polygonal cells with clear or light cytoplasm.
Intracytoplasmic lumina may be present. Mitotic figures
are infrequent. Pagetoid spread of cells between the surface
epithelial cells and the basement membrane may be present. The
differentiation between the two forms relates to the extent and
degree of distension of acini, but this is subject to intra- and
inter-observer variability. Because of this the term ‘lobular
neoplasia’ or ‘in situ lobular neoplasia’ was introduced, which
does not differentiate between ALH and LCIS.
A further system proposed is ‘lobular intraepithelial
neoplasia’37 with three subdivisions that relate to morphology
and predicted clinical outcome. This grading system requires
validation and has not been endorsed by the WHO at the
present time.38
‘Pleomorphic lobular carcinoma in situ’ is a more recently
described entity39e42 in which the cells show more marked
pleomorphism with larger nuclei. Central necrosis and calcification
may be present. Distinction from high-grade DCIS may,
on occasions, be difficult. Comedo-type necrosis can be present
in LCIS that has the typical cytological and architectural
features42 and, although atypical, this should not be diagnosed
as pleomorphic LCIS.
Other variants that have been described include endocrine,
apocrine, histiocytoid and signet ring lobular neoplasia,43 mainly
based on cytonuclear features.
Frequency
It is difficult to assess the frequency of lobular neoplasia since there
are no specific clinical, mammographic and macroscopic features.
It was considered to be present more frequently in the 40e50 age
group34 but the incidence in postmenopausal women is increasing,
probably due to detection by mammographic screening. The
incidence in otherwise benign biopsies is reported as between 0.5%
and 3.8%.34 44 It can be both multifocal and bilateral.
Immunohistochemical analysis
The characteristic alteration of lobular neoplasia is the lack of
expression of the cell adhesion molecule E-cadherin, which is
consistently present on the membranes of normal luminal
epithelial cells and is also lost in invasive lobular carcinoma
(ILC).45e47 Alterations are seen in both classical and pleomorphic
lobular neoplasia and immunohistochemical analysis of
E-cadherin can aid the diagnosis of solid in situ proliferations of
indeterminate nature.48e50 Lack of E-cadherin supports a diagnosis
of lobular neoplasia but, if a mixed pattern of staining is
present, then it should be reported as combined DCIS and
lobular neoplasia.50 Negative markers can, however, cause
problems in interpretation. p120 catenin is a member of the
cadherin-catenin complex that has a cytoplasmic staining
pattern in lobular neoplasia and has been found to be useful in
separating lobular neoplasia from low-grade DCIS.51
High molecular weight cytokeratins identified by the antibody
34bE12 have been reported to be consistently present in
lobular neoplasia and could be used to differentiate it from
DCIS,52 but a subsequent report considered staining to be an
artefact.53 This antibody should be used with caution.
Lobular neoplasia consistently shows positivity for ER and
PR, including the pleomorphic variant.41 47 HER2 amplification
and overexpression have been found in pleomorphic LCIS.41 42
Molecular studies
The CDH1 gene that encodes for E-cadherin is inactivated in
LCIS and ILC through both genetic (deletions and inactivating
mutations) and epigenetic (gene promoter methylation) mechanisms.
54e56 Concurrent mutations have been identified in LCIS
and ILC.46 Mutations have also been found in ALH.56
Comparative genomic hybridisation analysis has shown that
similar chromosomal changes are present in ALH and LCIS.57
Array comparative genomic hybridisation and mitochondrial
DNA analyses have confirmed similar genetic alterations
between matched lobular neoplasia and ILC.58 59
Overall, the molecular data provide strong evidence that
lobular neoplasia is a non-obligate precursor of ILC.
Differential diagnosis
The main problems relate to poor fixation in a surgical specimen,
differentiating low-grade solid DCIS from lobular neoplasia and
pleomorphic LCIS.
In larger surgical specimens poor fixation can lead to artefactual
discohesion within lobular units, resulting in an overdiagnosis
of LCIS. This is more of a problem in mastectomies
that have not been sliced sufficiently in the fresh state.
Differentiating low-grade solid DCIS from lobular neoplasia
can be difficult in both surgical specimens and core biopsies and
the use of E-cadherin staining can be particularly helpful.48e50
Coexistent low-grade DCIS and lobular neoplasia is not rare and
a mixed pattern of E-cadherin staining will be seen in these
circumstances. Cancerisation of lobules by DCIS can also cause
problems and again E-cadherin staining is of value.
Pleomorphic LCIS39e42 can be difficult to differentiate from
high-grade DCIS due to the degree of pleomorphism, the presence
of necrosis and calcification, and has most certainly been
classified as DCIS previously. Pleomorphic LCIS should be
considered if there is poor cellular cohesion along with these
other features. Again E-cadherin staining can be invaluable.
Since it is not possible reliably to assess the degree or extent of
lobular changes in core biopsies, providing that DCIS has been
excluded, the lesion should be reported as B3, of uncertain
malignant potential, and recorded as lobular neoplasia, with no
attempt to distinguish ALH and LCIS.
Significance
The significance of the diagnosis of lobular neoplasia in a needle
core biopsy relates to the risk of there being associated invasive
carcinoma (or DCIS) present at the time of the biopsy (see
Management). When identified within a surgical biopsy/excision
specimen, the questions of concern are different. These are: (1)
What is the risk of subsequent development of invasive carcinoma?
(2) Is lobular neoplasia a precursor lesion of invasive
disease? (3) Does lobular neoplasia affect the risk of recurrence
following breast conserving therapy?
It has been recognised for a long time that lobular neoplasia is
an indicator of increased risk of developing invasive cancer,34e36 44
776 J Clin Pathol 2012;65:771–785. doi:10.1136/jclinpath-2012-200733
Review
Downloaded from jcp.bmj.com on September 17, 2012 - Published by group.bmj.com
with the RR differing between ALH (3e53)60e62 and LCIS
(8e113).34 36 44 63 64 The increased risk has been found in some
reports to be evenly distributed between both breasts,34 64 65
although Page et al62 found invasive carcinoma to be three times
more likely to arise in the breast with ALH than the contralateral.
Collins et al66 found the risk of developing an invasive cancer to
be greater for premenopausal women with ALH than ADH,
whereas SEER data64 show an increased incidence after the
menopause.
The evidence that lobular neoplasia could be a non-obligate
precursor lesion comes from molecular and epidemiology studies.
As described above, the genetic profiles of ALH and LCIS are
similar to each other and to those of ILC.54e59 That there is
a greater risk of invasive cancer in the ipsilateral breast62 66 67 and
that the invasive cancer is more likely to be lobular than ductal35
67 also indicate that lobular neoplasia could be a non-obligate
precursor. Although there are no long-term follow-up studies of
pleomorphic LCIS, the small-size series that have been published
have shown a higher incidence of associated invasive disease,
usually ILC, which is suggestive of a precursor lesion.42
There is general agreement from published studies that the
presence and extent of lobular neoplasia, including presence at
the margins, does not increase the risk of recurrence following
breast conserving surgery,68e71 although the NSABP study69
excluded cases of pleomorphic LCIS (described as DLCIS).
Management
Needle core biopsy
Issues relating to lobular neoplasia in needle core biopsies have
recently been reviewed.72 73 The EUSOMA Working Group74
considered lobular neoplasia to be most frequently a coincidental
finding in a core biopsy and therefore advised that multidisciplinary
discussion was essential to determine management, as is
advocated by others.75 Diagnostic surgical excision of lobular
neoplasia has been advocated.76 A recent review of the literature
revealed an upgrade of 20% for LCIS and 13% for ALH to
carcinoma when excised.77 Concerns have been raised about
underestimation of cancer,77e81 even with stereotactic vacuumassisted
biopsy,79 and this has led to the recommendation of
diagnostic surgical excision for all such lesions from some
groups.73 It must be appreciated that much of the data is
retrospective and that not all studies have considered radiological-
pathological discordance as a factor resulting in upgrade, as
found by some.75 82 What is required are large prospective
studies with all factors included.
There is general agreement, although limited robust data, that
pleomorphic LCIS should be subjected to therapeutic excision; in
essence, treated as DCIS and therefore categorised as B5a on
needle core biopsy. In one series of 12 cases diagnosed on needle
core biopsy, ILC was found in 3 on subsequent excision.83 There
is a need for larger studies to confirm that this does represent
a more aggressive disease.
Surgical excisions
The presence and extent of lobular neoplasia in therapeutic
excisions should be reported as well as its presence at margins.
At present further surgery is not recommended if the invasive
cancer is excised.68e71
If lobular neoplasia only is seen in a diagnostic excision biopsy
and the clinical/radiologically relevant area has been excised,
then no further surgery is required.
If pleomorphic LCIS is found at excision margins then it
should be treated as would DCIS and further surgery undertaken
to obtain complete excision with appropriate margin of
clearance.
MICROMETASTASES AND ISOLATED TUMOUR CELLS (ITCS):
NHSBSP COORDINATING GROUP OVERVIEW AND
RECOMMENDATIONS
Lymph node status is recognised as being a major prognostic
factor for patients with breast cancer and forms part of staging
systems such as the TNM (tumour, lymph node, metastases)
classification system. That lymph node metastases could be
‘occult’ was recognised over 40 years ago,84 but their significance
is still debated. The introduction of sentinel lymph node biopsy
has raised further issues regarding the importance, or not, of
these ‘occult’ metastases. These relate to their definition; how
they are detected and measured; and their significance in
predicting involvement of non-sentinel lymph nodes and
prognosis.
Definitions
The term ‘occult’ metastasis has been defined as one missed by
initial histological examination and identified on subsequent
assessment, and also as a metastasis identified through additional
evaluation of paraffin-embedded lymph node blocks.85 It
has not been defined by size. Publications referring to occult
metastases are therefore not necessarily comparable with regard
to the size of these metastases. Some earlier studies did use
2.0 mm as the cut-off between micrometastasis and macrometastasis.
86e88 Based on the evidence produced from these, the
5th edition of the AJCC Cancer Staging Manual introduced the
definition of micrometastasis as being #2 mm (pN1a).89 The
sixth editions of the AJCC/UICC staging manuals90e92 further
defined micrometastases to be >0.2 mm but not >2.0 mm and
introduced the term isolated tumour cells (ITC) that are
#0.2 mm:
Key points and recommendations
< Due to problems in assessing the degree or extent of changes
in needle core biopsies, the terms ‘lobular neoplasia’ or ‘in situ
lobular neoplasia’ should be used rather than ‘atypical lobular
hyperplasia’ and ‘lobular carcinoma in situ’ (LCIS). In surgical
specimens, atypical lobular hyperplasia and LCIS should be
distinguished and recorded.
< Lack of expression of the cell adhesion molecule E-cadherin
can aid differentiation from low-grade solid DCIS.
< Pleomorphic LCIS exhibits more marked pleomorphism,
necrosis and calcification. It can be differentiated from DCIS
by lack of E-cadherin but should be managed as DCIS when
diagnosed on needle core biopsy (B5a) and if at surgical
excision margins.
< Lobular neoplasia is an indicator of increased risk of
developing invasive carcinoma, which is greater for LCIS.
< There is evidence that lobular neoplasia is a non-obligate
precursor lesion, based on similarities of genetic profiles with
ILC and greater risk of invasion in the ipsilateral breast.
< Finding of lobular neoplasia in needle core biopsy merits
multidisciplinary discussion and, for the majority, diagnostic
surgical excision.
< Presence of lobular neoplasia at surgical excision margins
should be recorded but does not require re-excision.
J Clin Pathol 2012;65:771–785. doi:10.1136/jclinpath-2012-200733 777
Review
Downloaded from jcp.bmj.com on September 17, 2012 - Published by group.bmj.com
Connolly,93 in discussing changes and problematic areas in the
AJCC Cancer Staging Manual, identified that there could be
differences in interpretation of ITCs in different situations
which would result in different classification of node status. The
European Working Group for Breast Screening Pathology (EWG)
undertook an evaluation of low volume lymph node involvement
in order to test reproducibility of these categories for
distinguishing ITCs and micrometastases.94 The first evaluation
of images from 50 lymph nodes showed only fair reproducibility,
better for micrometastases than for ITCs. Following discussions,
refinements to the definitions were proposed (box 1). The use of
these refinements improved reproducibility between participants.
When these criteria were used by another less specialised
group to categorise the same cases, consistency was achieved
with the experts95 but the level of consistency was still
moderate. A subsequent study to improve interobserver
reproducibility was undertaken led by Turner,96 in which six
experienced breast pathologists examined digital images of
small volume nodal metastases before and after studying
a training programme (criteria in box 2). The EWG then applied
the two interpretations of the TNM definitions and found
that the Turner system had better reproducibility but it may
underestimate the rate of non-sentinel node involvement.97
The evidence suggests that there are problems with using
these definitions, particularly when discriminating between
ITCs and micrometastases which will affect interpretation
of their significance. The TNM definitions of low volume
nodal metastases need to be better formulated with visual
information.
Detection
The three areas that have been considered have been the extent
to which lymph nodes should be examined histologically, the
use of IHC and, to a lesser extent, the use of reverse transcriptase
PCR to identify micrometastases and ITCs.
Histological examination of lymph nodes
How extensively to analyse a lymph node has become a question
raised frequently with the increasing use of the sentinel lymph
node biopsy technique.
There are differences in relation to the initial slicing of the
node; the American Society of Clinical Oncology (ASCO)
guidelines for sentinel lymph node biopsy in early breast
cancer98 recommend slicing through the long axis whereas
NHSBSS Pathology Reporting of Breast Disease99 recommends
slicing the node perpendicular to the long axis. Both advocate
thin slicing and embedding all pieces. When sentinel nodes are
sliced at 2 mm intervals and totally embedded, the probability of
identifying all metastases >2.0 mm is high.85 Cserni100 used
a geometrical model of sentinel lymph node histopathology and
found that sections 1 mm apart should identify metastases
>2 mm. To identify micrometastases, step sectioning with levels
of 250 mm or 200 mm would be required.
A review of the literature101 on the value of using step
sectioning and IHC (see below) found upstaging to be reported
Box 2 Turner study classification rules (from Cserni
et al97)
< Isolated tumour cells and micrometastases are distinguished
by the size of the largest tumour cell cluster, regardless of
microanatomical location within or adjacent to the lymph node
and regardless of the number of clusters or single cells.
< A cluster is a confluent focus of tumour cells touching other
tumour cells.
< Clusters or cells separated by a single benign cell or a spatial
gap are measured as separate clusters, except when
fibroblastic reaction has caused the separation.
< Any lesion composed of non-cohesive cells or clusters not
>0.2 mm represents isolated tumour cells.
< Any lesion with largest cohesive cellular lesion >0.2 mm but
not >2.0 mm represents micrometastasis.
< Mitotic activity is not considered.
< For borderline or indeterminate findings at the two ends of the
spectrum, the lower stage category should be selected.
Micrometastases pN1mi, >0.2 mm, none >2 mm.
May have histological evidence of
malignant activity (proliferation/stromal
reaction) but not a requirement.
Isolated tumour cells (ITC) Single tumour cells or small clusters of
cells no more than 0.2 mm in greatest
dimension. Usually detected by immunohistochemistry
or molecular methods,
verified by H&E section. Do not typically
show proliferation/stromal reaction or
penetration of vascular or lymphatic sinus
walls.
pN0dno metastasis histologically.
pN0 (i+) isolated tumour cells.
Box 1 European Working Group study classification rules
(from Cserni et al97)
< Capsular (including intravascular) lesions considered nodal,
and any lesion with nodal and simultaneous extranodal
involvement considered nodal, but lesions outside the node
(afferent lymphatics, perinodal fat) not recorded as nodal
involvement.
< Tumour cells (multiple) localised in parenchyma considered
micrometastasis even if <0 .2=".2" mm="mm" no="no" or="or" p="p" proliferation="proliferation" with="with">stromal reaction.
< Tumour cells localised in vessels or sinuses: if #0.2 mm
isolated tumour cells, if >0.2e2 mm micrometastasis.
< If multiple foci, only largest should be considered.
< Single tumour cells or clusters arranged in a continuous
manner or separated by 2e5 cells should be considered and
measured as one focus and characterised by largest
dimension.
< Cells or clusters arranged in a discontinuous manner and
dispersed homogenously in a definable part of the lymph node
should be considered and measured as one focus.
< Cells, clusters or foci (as defined above) arranged in
a discontinuous manner and dispersed unevenly should be
considered as one if the distance between the clusters or foci
is smaller than the smallest cluster or focus.
< Cells, clusters or foci (as defined above) arranged in
a discontinuous manner and dispersed unevenly should be
considered as distinct and multiple if the distance between
the unevenly distributed tumour cells, clusters or foci is
greater than the smaller cluster or focus and should be
characterised by the largest cluster or focus.
< In cases of doubt, the lower category (isolated tumour cell)
should be given.
778 J Clin Pathol 2012;65:771–785. doi:10.1136/jclinpath-2012-200733
Review
Downloaded from jcp.bmj.com on September 17, 2012 - Published by group.bmj.com
after the former to range from 4.8%102 to 30.7%.103 However, it
was difficult to assess in various studies whether this referred to
macrometastasis or micrometastasis.
Immunohistochemistry (IHC)
Cytokeratin antibodies (eg, Cam5.2, MNF 116, Pan cytokeratin;
AE1/AE3) have been used in many studies to aid the detection
of metastases (macrometastases and micrometastases, ITCs),
often in combination with step sectioning and on multiple
levels in cases with negative H&E findings. Reviews of the
literature101 104 indicate IHC results in upstaging in 2.6e19% of
cases. One study105 found IHC to detect metastases in 4%, 2%
being micrometastases (detectable on H&E section on review)
and 2% being ITCs, and concluded it was of limited value.
Concerns about interpretation of IHC were raised by Roberts
et al106 who found that, as tumour cell numbers decreased, the
ability of pathologists to recognise them also decreased. Other
concerns have been raised about IHC-only detected tumour cells
in lymph nodes.107 In a review of 4066 cases of sentinel nodes
positive only on IHC, there was no association with vascular
invasion, tumour size, tumour type and location but there was
an association with method of biopsy, the frequency increasing
in relation to the degree of manipulation.
Molecular analysis
The predominant method has been reverse transcriptase PCR for
a range of markers (cytokeratin 19, 20, CEA, MUC-1, maspin,
mammoglobin) using either frozen sections or half the sentinel
node. One of the largest series with 123 patients and 146 nodes
by Manzotti et al108 had a high sensitivity but lower specificity
for any marker, with a higher specificity for two of CK19,
maspin or mammoglobin. Molecular analysis has recently been
developed commercially for the intraoperative assessment of
metastases in sentinel lymph nodes109e111 but has not been
evaluated extensively.
Significance
There are two topics of importance: (1) whether micrometastases
and ITCs predict the involvement of non-sentinel
axillary lymph nodes and (2) the prognostic significance of
micrometastases and ITCs.
Predicting non-sentinel lymph node metastasis
The aim of the sentinel node procedure is to reduce the
frequency of axillary node surgery in node negative women and
so reduce morbidity. Identification of sentinel node involvement
is considered an indication for further axillary surgery or radiotherapy,
but the question is whether such treatment is required
if only a micrometastasis is present in the sentinel node. Cserni
et al112 undertook a literature review and meta-analysis of 25
studies published up to 2003 that considered non-sentinel node
metastases associated with sentinel node micrometastasis. They
concluded that the risk of non-sentinel node metastasis was
around 10e15%, lower if sentinel node involvement was
detected by IHC. Viale et al113 found differences in frequency of
non-sentinel node metastases in relation to the size of the
micrometastasis, being less if <1 factors="factors" have="have" mm.="mm." other="other" p="p" that="that">been found to relate to non-sentinel node metastases in the
presence of sentinel node micrometastasis include tumour
size,114e116 grade115 and vascular invasion.116 Since the metaanalysis
undertaken by Cserni et al,112 further publications have
reported differing frequencies in the incidence of non-sentinel
node metastases. In tumours 15 mm or less, no further
involvement was found for cases with ITCs, with 12% of cases
with micrometastases having non-sentinel node involvement.117
Tumour size and type were found to be factors predicting noninvolvement
in the study by Houvenaeghel et al.118 Similar
frequencies of involvement for cases with micrometastases have
been reported in two papers from Holland (19%, 20%),119 120
although there were differences for ITCs (7%, 13%). In a study
confined to ITC positive sentinel lymph nodes, a very low
incidence of non-sentinel node metastases was found and no
axillary recurrence if axillary surgery had not been performed.121
There is sufficient evidence to support the avoidance of
axillary node dissection in cases with ITCs, but there is no
agreement with regard to the further management of the axilla
if micrometastases are identified.
Prognostic significance of micrometastases and isolated tumour
cells (ITCs)
Many of the studies undertaken (reviewed by Cserni et al101)
have considered occult metastases with only a small number
commenting specifically on micrometastasis. Different combinations
of H&E 6 step sectioning 6 IHC have been used.
There is no strong evidence for a survival disadvantage. Some
studies suggest that there is, but within these there are variations
in relation to disease-free and overall survival and
different patient groups. A study with a long follow-up that
considered occult metastases found no difference in survival for
patients with and without these,122 and emphasised the need
for multivariate analysis in such studies. Using data taken from
a cancer registry for a 25-year period, Kuijt et al123 found on
multivariate analysis that patients with micrometastases had
a significantly worse survival than those without, but there
was no pathological review of lymph nodes. The same applies
in two studies based on data from SEER which found the
prognosis for micrometastatic disease intermediate between
N0 and N1124 125 and concluded that a prospective study is
required. A re-evaluation of node negative cases with 20-year
follow-up using IHC identified 23% of cases with metastatic
disease, predominantly ITCs.126 The prognosis of those with
micrometastatic disease was poorer than node negative cases,
which was more apparent after 10 years. Another centre
examining node negative cases after 8 years of follow-up found
13% of cases with micrometastatic disease but there was no
association with outcome.127
USE OF BASAL/MYOEPITHELIAL MARKERS IN BREAST
PATHOLOGY: NHSBSP COORDINATING GROUP OVERVIEW AND
RECOMMENDATIONS
The increasing use of needle core biopsy for non-operative
diagnosis and the nature of lesions identified mammographically
can result in diagnostic difficulties. Interpretation may be helped
by the use of IHC to detect basal/myoepithelial markers. A
recently circulated questionnaire showed that there were variations
in the type and frequency of markers used diagnostically in
the UK and Ireland. This paper summarises current knowledge
of the various markers, discusses the diagnostic areas where they
are of value and makes recommendations for a suitable diagnostic
panel.
Basal/myoepithelial markers
The myoepithelial cell has traditionally been distinguished from
luminal epithelial cells by the presence of smooth muscle fibres.
However, many more proteins have now been identified that are
expressed in myoepithelial cells and these fall into three groups
(smooth muscle-related, cytokeratins and others).
J Clin Pathol 2012;65:771–785. doi:10.1136/jclinpath-2012-200733 779
Review
Downloaded from jcp.bmj.com on September 17, 2012 - Published by group.bmj.com
Smooth muscle-related
< Smooth muscle actin (SMA): antibodies detect actin microfilaments;
good sensitivity but specificity poor since will
detect myofibroblasts.128
< Smooth muscle myosin heavy chain (SMMHC): structural
component of smooth muscle myosin. SM2 isoform is
expressed in breast myoepithelial cells. Sensitivity and specificity
are high with SMMHC being present in periductal and
periacinar myoepithelial cells and not stromalmyofibroblasts,129
although sensitivity reported to be slightly lower than SMA.128
< Calponin: 34 kD polypeptide, modulates actomyosin ATPase
activity. Excellent sensitivity but present in a subset of
myofibroblasts.128 129
< H-caldesmon: SMA binding protein. Only detectable
in myoepithelial cells of ducts but not found in
myofibroblasts.129 130
Cytokeratins
< Myoepithelial cells express cytokeratins (CK) characteristic of
basal layer of stratified epithelium: CK5, CK10, CK14 and
CK17.131 132
< CK5/6 in normal breast can be detected in both myoepithelial
and luminal epithelial cells.133
< The antibody 34b12 recognises CKs 1, 5, 10 and 14 but is not
specific for myoepithelial cells.134
Other markers
< S100: has low specificity since detectable in epithelial cells.135
< CD10 (Common Acute Lymphoblastic Leukaemia Antigen
(CALLA)): endopeptide expressed in myoepithelial cells.136
Antibodies now available that work on fixed tissue; high
sensitivity in normal breast.137
< P cadherin: cell adhesion molecule with high sensitivity for
myoepithelial cells in normal breast, no reactivity with
myofibroblasts.138
< P63-p53 homologue: differs from other markers in being
nuclear139 with high sensitivity for myoepithelial cells.140
< 14-3-3s: protein associated with apoptosis and cell cycle
control, present in myoepithelial cells in normal breast.141
< Maspin: serine protease inhibitor present in myoepithelial
cells142 with both cytoplasmic and nuclear reactivity.
Diagnostic areas
Radial scar versus tubular carcinoma
Highly sensitive and specific markers are required due to
myoepithelial cells in central parts of radial scars being attenuated,
plus the presence of myofibroblasts. The requirements
favour SMMHC and p63 as markers of choice. SMA and, to
a lesser extent, calponin will be present in myofibroblasts.
Papillary lesions
IHC can reduce the variability in reporting of core biopsies of
papillary lesions, particularly in relation to B3 and B4.143 Intraduct
papillomas have a complete layer of myoepithelial cells,
whether detected by SMMHC, calponin, p63 and CK5/6144 or
SMA, p63, CD10 and CK14.145 However, findings for intracystic
papillary carcinomas differ, with Collins et al144 failing to detect
any myoepithelial cells and Tse et al145 finding them to be
scattered and discontinuous. p63 had the highest sensitivity,
CK14 was also present in florid hyperplasia, SMA showed
stromal staining and CD10 epithelial and stromal staining.
Hyperplasia versus atypia versus DCIS
High molecular weight basal cytokeratins are of use particularly
in interpretation of core biopsies enabling a B2 rather than a B3
diagnosis. Myoepithelial cells form a useful internal positive
control. Hyperplasia of usual type and hyperplasia in papillomas
show reactivity for CK5/6133 146 147 and CK14146 whereas
atypical hyperplasia and DCIS epithelium do not.
Non-invasive versus invasive carcinoma
Myoepithelial cell markers can be of value in assessing whether or
not invasion is present, but myofibroblast reactivity, discontinuity
of staining of myoepithelial cells and staining of vascular smooth
muscle cells can cause problems in interpretation. Discontinuous
staining can occur with p63,140 148 but it has greater sensitivity
and specificity. SMMHC has been found to be more sensitive than
CD10 in assessing invasion149 but is in vascular smooth
muscle cells. Owing to the advantages and disadvantages, it is
better to use two myoepithelial markers that complement one
anotherdfor example, p63 and SMMHC or calponin.
Myoepithelial tumours
SMA, CK5/6, CK14, p63 and other markers are all suitable for
analysing possible (adeno) myoepithelial tumours, both benign
and malignant.150
Other lesions
Myoepithelial markers can aid differentiation of adenoid cystic
carcinoma from collagenous spherulosis and cribriform carcinoma.
Basal-like carcinomas
There is increasing interest in basal-like breast carcinomas, partly
due to the finding that BRCA1-related breast cancers can have
these features151 and to their poorer response to anthracycline
Key points and recommendations
Despite the extensive literature, there is still a lot of debate about
the importance and significance of micrometastases and isolated
tumour cells (ITCs), particularly with increasing use of sentinel
node biopsy. This impacts on factors such as the routine
pathological handling of sentinel nodes.
< If a micrometastasis is identified in a single H&E section,
levels should be cut to check whether it remains a micrometastasis
or becomes a macrometastasis (pN1mi to pN1).
< Sentinel lymph nodes should be sliced at 1e2 mm with all pieces
embedded to ensure a full face H&E section. This is the minimum
requirement. Step sectioning at 200e250 mm can be undertaken
but is not mandatory. It would be appropriate for centres
undertaking detailed clinical studies of sentinel lymph nodes.
< There is no good evidence for the routine use of
immunohistochemistry in evaluating sentinel lymph nodes; it
could be of value for centres undertaking detailed clinical
studies of sentinel lymph nodes.
< DefinitionsdAJCC/UICC amended definitions to be followed;
discrimination between micrometastases and ITCs can be
aided by using criteria from the European Working Group and
Turner studies.
< Since there are variations in the incidence of non-sentinel
node metastases in the presence of sentinel node micrometastasis,
axillary surgery/radiotherapy is considered appropriate
but is not indicated for ITCs.
< There is no strong evidence for an effect of micrometastases
on survival.
Further data are required in several areas to assess the significance
and importance of micrometastases and isolated tumour cells.
780 J Clin Pathol 2012;65:771–785. doi:10.1136/jclinpath-2012-200733
Review
Downloaded from jcp.bmj.com on September 17, 2012 - Published by group.bmj.com
chemotherapy.152 There is no consensus as to the definition of
basal-like, which probably reflects the view that they are
a heterogeneous group of cancers. Nielsen et al153 consider them to
express CK5/6, HER-1 (EGFR) and/or c-kit and lack ER and HER2.
Others have used CK14154 or P-cadherin155 as a basal marker to
identify these tumours. Rakha et al156 have proposed that basallike
carcinomas can be defined based on the expression of basal
cytokeratins (CK5/6 and CK14) irrespective of other markers.
OESTROGEN RECEPTOR ASSESSMENT IN DUCTAL CARCINOMA
IN SITU (DCIS)
The response of invasive breast cancer to endocrine therapy is
well recognised as being related to the hormone receptor status
of the tumour.157 It is therefore logical to investigate the role of
hormone receptors in determining the role of endocrine therapy
in the management of DCIS. However, to date, there remain
issues with regard to the latter and with the assessment of
hormone receptors.
Evidence from clinical trials
The value of tamoxifen in the management of DCIS has been
reported in two randomised studies:
1. The NSABP B-24 trial of the use of tamoxifen and
radiotherapy in DCIS showed a benefit from endocrine
therapy, primarily due to a reduction of ipsilateral recurrent
invasive carcinoma.157 158 The 7-year risk of local recurrence
in the treated breast after lumpectomy plus radiation was
reduced from 11.1% without tamoxifen to 7.7% with
tamoxifen (p¼0.02). The risk of all breast cancer events
(ipsilateral plus contralateral) was reduced from 16.9% to
10.0% (p¼0.0003). Allred and colleagues retrospectively
reviewed 628 cases from this trial and demonstrated that
the benefit was confined to those patients whose tumours
were ER positive; this was defined as 10% or more cells
staining.159 However, it is not clear whether there was
a difference in ipsilateral and/or contralateral recurrence with
regard to ER status of the primary disease process as the data
are available in abstract form only. It is also not clear how the
figure of 10% was reached and, in particular, whether
modelling was carried out to determine a cut-off value or
range that best correlated with therapeutic benefit. The
review included a mixture of both locally reported ER
and central assays and concern was raised about possible
false negative results from laboratories using diverse nonstandardised
assays. With the exception of the abstract
referred to above, the study has not been published.
2. The UK/ANZ (UKCCCR) Trial160 comparing radiotherapy
and tamoxifen in the treatment of DCIS showed after
53 months of follow-up a non-significant reduction in all
breast events with the addition of tamoxifen but a RR
reduction of 22%. It should be noted that the follow-up
period in the UK/ANZ trial at the time of reporting was
shorter than NSABP B-24 (4.4 vs 6.9 years) and receptor
status of the DCIS was not measured prospectively in this
trial. In addition, a high proportion of cases were of high
cytonuclear grade; a relatively high proportion are therefore
likely to be ER negative which will have influenced the effect
of hormone treatment in this series (S E Pinder, personal
communication).
A further study, the NSABP-P1 trial of tamoxifen for the
prevention of breast cancer, studied 13 000 at-risk women.161
This cohort did not include patients with DCIS but illustrates
the potential benefit of endocrine therapy in the prevention of
breast cancer events. Invasive and non-invasive carcinomas were
reduced by 50% at 69 months follow-up. There was a threefold
increase in thromboembolic events, a marginal increase in
cataracts and a non-significant increase in strokes.
Current guidance and options
A cut-off point for positive versus negative for ER in DCIS was
set at <5 an="an" as="as" cells="cells" clinical="clinical" criterion="criterion" entry="entry" for="for" p="p" staining="staining" two="two" uk="uk">trials of endocrine therapy in DCIS (IBIS II and DCIS II, the
latter discontinued). The UK breast screening guidelines162
recommend the use of <5 and="and" cells="cells" dcis="dcis" for="for" p="p" staining="staining" the="the" use="use">of the Allred score for invasive cancers. A cut-off of Allred score 3
has been used in a large study of pure DCIS examining the
distribution of various different phenotypes162 and is the same
as that applied to invasive breast carcinoma.157 162 It would be
illogical to adopt a different scoring system from those already
in use for invasive disease unless sound evidence to the contrary
emerged. Furthermore, there is strong evidence that the ER
status of invasive breast cancer parallels that of associated DCIS
in individual cases.163 Individual practitioners benefit from
familiarity with a particular scoring technique and clinical
colleagues require consistency in the approach to reporting. The
three scoring methods currently in use worldwide are: (1)
a simple percentage of cells staining without reference to
intensity; (2) the histochemical ‘H’ score which is the sum of the
percentage of cells with three levels of staining, giving a range of
0e300164; and (3) Allred scoring (replacing previous ‘Quick
Score’) which is the sum of an intensity score of 1e3 and the
percentage of cells staining banded in a non-linear fashion using
a score of 1e5 giving a final score between 2 and 8.165
Allred scoring and the histochemical score provide similar
information about receptor status as both include an assessment
of intensity as well as percentage of cell nuclei staining. Simple
percentage scoring gives no information about staining intensity
although low proportion scores tend to go hand in hand with
low intensity scoring with ER scoring, as opposed to PR
immunoreactivity.
There is no evidence base to support a preference for
a particular scoring technique. However, there is evidence for
assessment of proportion and intensity staining in invasive
breast cancer and, as such evaluation is routine practice, it would
be logical to mirror this practice in the reporting of DCIS.
Evidence from the Sloane Project,166 a UK audit of screendetected
DCIS and atypical hyperplasias of the breast, points to
Recommendations for a diagnostic panel
< It is important not to rely on just one marker.
< Smooth muscle myosin heavy chain (SMMHC) or calponin are
sensitive and specific cytoplasmic myoepithelial markers.
Smooth muscle actin can be used but care has to be taken in
interpretation in the presence of myofibroblasts.
< P63 is a sensitive and specific nuclear myoepithelial marker
but staining can be discontinuous, so use of a cytoplasmic
myoepithelial marker such as SMMHC or calponin as well will
aid interpretation.
< When trying to identify the presence/absence of myoepithelial
cells, use p63 and SMMHC/calponin.
< CK5/6 is of value in several diagnostic situations such as
analysis of hyperplasias or basal-like breast cancers.
J Clin Pathol 2012;65:771–785. doi:10.1136/jclinpath-2012-200733 781
Review
Downloaded from jcp.bmj.com on September 17, 2012 - Published by group.bmj.com
considerable confusion in this area among pathologists in their
reporting practice of ER status in DCIS. A wide variation is seen
between and within laboratories of the scoring method used and
also of the cut-off point for positive versus negative results. In
this audit, 40% of reporting laboratories used one method and
the same cut-off criterion throughout, 19% used one method but
different cut-off criteria for different cases, 15% of laboratories
used two methods for different cases with equivalent cut-off
points for each method while 25% of laboratories used two
methods and non-equivalent cut-off points from case to case.
Because there is such uncertainty about what cut-off is
therapeutically relevant (if any) in the pharmacological
management of DCIS and possibly as a more general prognostic
marker for this disease process, it would be sensible to record the
percentage of cells staining and the average intensity in the duct
profiles available and correlate this information with clinical
outcomes.
Patterns of ER expression in DCIS
From the data gathered by the Sloane Project, intermediate and
low-grade DCIS are almost invariably ER positive whereas
high-grade DCIS is positive in 69% of cases (table 2).
Baqai and Shousha reviewed 60 cases of DCIS, 56 pure and
four with associated microinvasion. DCIS was assessed in terms
of grade, architecture and, among other things, ER positivity
was defined as disease with >10% of cells showing dark brown
nuclear staining (Allred score >4, Histoscore >20). The grade
distribution seen was 52% high, 38% intermediate and 10% low,
which is very similar to the distribution among the Sloane
Project cases.168 There was a strong association between ER
negativity and high nuclear grade. The number of cases in this
study was small and the cut-off point was set relatively high. It
is a pity that the opportunity to model different cut-off points
against grade was not taken.
Ottesen et al169 showed in a series of 133 cases of DCIS that
one-third displayed 5% or less staining for ER, the overwhelming
majority of which were of large nuclear size. In this series,
perhaps unusually, most cases were either strongly positive or
negative.
Basal phenotype DCIS, seen in approximately 6% of
high-grade lesions, is commonly negative for ER, PR and
Her2.162 170 171
Future directions
It is likely that the IBIS II trial will give invaluable information
about the validity of setting a positive/negative cut-off point at
5% or 10% for ER. Obviously there will be limited information
about the relevance of staining intensity unless Allred scores are
used and these are not obligatory. The Sloane Project, given its
long-term goals of prolonged follow-up of DCIS patients, will
also provide information about a range of cut-off points because
of the variability that has already been demonstrated in the first
cohort of patients reviewed.167 Because data are not available to
give guidance on the clinical relevance of different proportion
and intensity scores, it is recommended as a minimum to record
both when reporting the hormone receptor status of DCIS. This
will enable Histoscores and/or Allred scores to be derived
without loss of the original scoring information.
There remains considerable scope for modelling ER cut-off
points in DCIS against clinical outcomes in large cohorts of
patients but, until such time as that is possible, it seems
reasonable to apply the limited evidence available at the present
time and use the currently accepted cut-off points to guide the
endocrine management of patients with DCIS as for reporting
ER status in invasive breast cancer.162
Acknowledgements The authors acknowledge the members of the UK National
Coordinating Committee for Breast Pathology.
Contributors These are working documents aimed to established early guidance on
emerging areas of diagnostic breast pathology.
Competing interests None.
Provenance and peer review Commissioned; internally peer reviewed.
REFERENCES
1. Fraser JL, Raza S, Chorny K, et al. Columnar alteration with prominent apical
snouts and secretions: a spectrum of changes frequently present in breast biopsies
performed for microcalcifications. Am J Surg Pathol 1998;22:1521e7.
2. Ishihara A, Kobayashi TK. Infarcted intraductal papilloma of the breast: cytologic
features with stage of infarction. Diagn Cytopathol 2006;34:373e6.
3. Cameselle-Teijeiro J, Abdulkader I, Barreiro-Morandeira F, et al. Breast tumor
resembling the tall cell variant of papillary thyroid carcinoma: a case report. Int J
Surg Pathol 2006;14:79e84.
Table 2 Grade of DCIS and ER status: Sloane Project data based on
1684 cases
Grade High (%) Intermediate (%) Low (%) Unknown (%) Total (%)
994 (59) 490 (29) 181 (11) 19 (1) 1684
ER positive 325 (69) 197 (94) 79 (99) 5 606 (79)
ER negative 148 (31) 12 (6) 1 (1) 161 (21)
Unknown 521 281 101 14
DCIS, ductal carcinoma in situ; ER, oestrogen receptor.
Key points and recommendations
< >80% of DCIS is ER positive and there is a body of evidence
indicating a therapeutic benefit for endocrine treatment in
selected ER positive patients.
< Previously a cut-off point of 5% of cells staining has been
recommended to define positive and negative. It is recommended
that both proportion of cells staining and intensity
should be recorded.
Take-home messages
< Columnar cell lesions may show a spectrum of abnormalities
from simple metaplasia to ductal carcinoma in situ.
< Lobular neoplasia or in situ lobular neoplasia are the preferred
terms for core needle biopsy reports whereas the terms
atypical lobular hyperplasia or lobular carcinoma in situ should
be specified in excisions.
< Metastatic tumour clusters in lymph nodes <0 .2=".2" are="are" mm="mm" p="p">designated isolated tumour cells. Discontinuous clusters
should be measured as one focus if in a definable part of
a lymph node. Unevenly dispersed clustered should be
characterised by the largest cluster.
< When using basal/myoepithelial markers do not rely on
a single marker. There is often mismatch between individual
marker’s staining profiles.
< ER scoring for DCIS should follow practice in invasive disease
recording both proportion and intensity of cells staining using,
for example, a Histoscore or Allred type system.
782 J Clin Pathol 2012;65:771–785. doi:10.1136/jclinpath-2012-200733
Review
Downloaded from jcp.bmj.com on September 17, 2012 - Published by group.bmj.com
4. Tremblay G, Deschenes J, Alpert L, et al. Overexpression of estrogen receptors in
columnar cell change and in unfolding breast lobules. Breast J 2005;11:326e32.
5. Vincent-Salomon A. [Columnar lesions: a frequent diagnosis in breast pathology!]
(In French). Ann Pathol 2003;23:593e6.
6. Schnitt SJ, Vincent-Salomon A. Columnar cell lesions of the breast. Adv Anat
Pathol 2003;10:113e24.
7. Tavassoli FA, Devilee P. Pathology and Genetics of Tumours of the Breast and
Female Genital Tract. Lyon: IARC Press, 2003.
8. Page D, Anderson T. Diagnostic Histopathology of the Breast. Edinburgh: Churchill
Livingstone, 1987.
9. Rosen PP. Columnar cell hyperplasia is associated with lobular carcinoma in situ
and tubular carcinoma. Am J Surg Pathol 1999;23:1561.
10. Rosen PP. Rosen’s Breast Pathology. 2nd edn. Philadelphia: Lippincott Williams &
Wilkins, 2001.
11. Foote FW, Stewart FW. Comparative studies of cancerous versus noncancerous
breast I. Basic morphologic characteristics. Ann Surg 1945;121:6e53.
12. Goldstein NS, O’Malley BA. Cancerization of small ectatic ducts of the breast by
ductal carcinoma in situ cells with apocrine snouts: a lesion associated with tubular
carcinoma. Am J Clin Pathol 1997;107:561e6.
13. Sasse F. U¨ber Cysten und cystiche Tumoren der Mamma. Arch Klin Chir
1897;54:1e54.
14. Schimmelbusch G. Das Cystadenom der Mamma. Arch Klin Chir 1892;44:117e34.
15. Oyama T, Maluf H, Koerner F. Atypical cystic lobules: an early stage in the
formation of low-grade ductal carcinoma in situ. Virchows Arch 1999;435:413e21.
16. Bonser G, Dossett JA, Jull JW. Human and Experimental Breast Cancer. London:
Pitman Medical, 1961.
17. Wellings SR, Jensen HM, Marcum RG. Atlas of subgross pathology of the human
breast with special reference to possible cancerous lesions. J Natl Cancer Inst
1975;55:231e73.
18. Kusama R, Fujimori M, Matsuyama I, et al. Clinicopathological characteristics of
atypical cystic duct (ACD) of the breast: assessment of ACD as a precancerous
lesion. Pathol Int 2000;50:793e800.
19. Moinfar F, Man YG, Bratthauer GL, et al. Genetic abnormalities in mammary ductal
intraepithelial neoplasia- at type (“clinging ductal carcinoma in situ”): a simulator of
normal mammary epithelium. Cancer 2000;88:2072e81.
20. Allred DC, Mohsin SK. Biological features of premalignant disease in the human
breast. J Mammary Gland Biol Neoplasia 2000;5:351e64.
21. Viale G. Histopathology of primary breast cancer 2005. Breast 2005;14:487e92.
22. Brogi E, Oyama T, Koerner FC. Atypical cystic lobules in patients with lobular
neoplasia. Int J Surg Pathol 2001;9:201e6.
23. McLaren BK, Gobbi H, Schuyler PA, et al. Immunohistochemical expression of
estrogen receptor in enlarged lobular units with columnar alteration in
benign breast biopsies: a nested case-control study. Am J Surg Pathol
2005;29:105e8.
24. Bratthauer GL, Tavassoli FA. Assessment of lesions coexisting with various
grades of ductal intraepithelial neoplasia of the breast. Virchows Arch
2004;444:340e4.
25. The Mammary Gland: Development, Regulation, and Function. New York, NY:
Plenum Press, 1987.
26. Van Poznak C, Cross SS, Saggese M, et al. Expression of osteoprotegerin (OPG),
TNF related apoptosis inducing ligand (TRAIL), and receptor activator of nuclear
factor kappaB ligand (RANKL) in human breast tumours. J Clin Pathol
2006;59:56e63.
27. Otterbach F, Bankfalvi A, Bergner S, et al. Cytokeratin 5/6 immunohistochemistry
assists the differential diagnosis of atypical proliferations of the breast.
Histopathology 2000;37:232e40.
28. Dabbs DJ, Carter G, Fudge M, et al. Molecular alterations in columnar cell lesions
of the breast. Mod Pathol 2006;19:344e9.
29. Eusebi V, Feudale E, Foschini MP, et al. Long-term follow-up of in situ carcinoma of
the breast. Semin Diagn Pathol 1994;11:223e35.
30. Simpson PT, Gale T, Reis-Filho JS, et al. Columnar cell lesions of the breast: the
missing link in breast cancer progression? A morphological and molecular analysis.
Am J Surg Pathol 2005;29:734e46.
31. Tan PH, Ho BC, Selvarajan S, et al. Pathological diagnosis of columnar cell
lesions of the breast: are there issues of reproducibility? J Clin Pathol
2005;58:705e9.
32. O’Malley FP, Mohsin SK, Badve S, et al. Interobserver reproducibility in the
diagnosis of at epithelial atypia of the breast. Mod Pathol 2006;19:172e9.
33. Foote FW Jr, Stewart FW. Lobular carcinoma in situ. A rare form of mammary
cancer. Am J Pathol 1941;17:491e6.
34. Haagensen CD, Lane N, Lattes R, et al. Lobular neoplasia (so-called lobular
carcinoma in situ) of the breast. Cancer 1978;42:737e69.
35. Wheeler JE, Enterline HT, Roseman JM, et al. Lobular carcinoma in situ of the
breast. Long-term followup. Cancer 1974;34:554e63.
36. Andersen JA. Lobular carcinoma in situ. A long-term follow-up in 52 cases. Acta
Pathol Microbiol Scand A 1974;82:519e33.
37. Bratthauer GL, Tavassoli FA. Lobular intraepithelial neoplasia: previously
unexplored aspects assessed in 775 cases and their clinical implications. Virchows
Arch 2002;440:134e8.
38. Lobular neoplasia. In: Tavassoli FA, Devilee P, eds. Pathology and Genetics of
Tumours of the Breast and Female Genital Organs. World Health Organization
Classification of Tumours. Lyon: IARC Press, 2003:60e2.
39. Eusebi V, Magalhaes F, Azzopardi JG. Pleomorphic lobular carcinoma of the breast:
an aggressive tumor showing apocrine differentiation. Hum Pathol
1992;23:655e62.
40. Sneige N, Want J, Baker BA, et al. Clinical, histopathologic and biologic features of
pleomorphic lobular (ductal-lobular) carcinoma in situ of the breast: a report of 24
cases. Mod Pathol 2002;15:1044e50.
41. Reis-Filho JS, Simpson PT, Jones C, et al. Pleomorphic lobular carcinoma of the
breast: role of comprehensive molecular pathology in characterization of an entity.
J Pathol 2005;207:1e13.
42. Fadare O, Dadmanesh F, Alvarado-Cabrero I, et al. Lobular intraepithelial neoplasia
[lobular carcinoma in situ] with comedo-type necrosis. Am J Surg Pathol
2006;30:1445e53.
43. Koerner F, Maluf H. Uncommon morphologic patterns of lobular neoplasia. Ann
Diagn Pathol 1999;3:249e59.
44. Page DL, Kidd TE Jr, Dupont WD, et al. Lobular neoplasia of the breast: higher risk
for subsequent invasive cancer predicted by more extensive disease. Hum Pathol
1991;22:1232e9.
45. De Leuuw WJ, Berx G, Vos CB, et al. Simultaneous loss of E-cadherin and
catenins in invasive lobular breast cancer and lobular carcinoma in situ. J Pathol
1997;183:404e11.
46. Vos CB, Cleton-Jansen AM, Berx G, et al. E-cadherin inactivation in lobular
carcinoma in situ of the breast: an early event in tumorigenesis. Br J Cancer
1997;76:1131e3.
47. Simpson PT, Gale T, Fulford LG, et al. The diagnosis and management of
preinvasive breast disease: pathology of atypical lobular hyperplasia and lobular
carcinoma in situ. Breast Cancer Res 2003;5:258e62.
48. Maluf HM. Differential diagnosis of solid carcinoma in situ. Semin Diagn Pathol
2001;25:237e44.
49. Maluf HM, Swanson PE, Koerner FC. Solid low-grade in situ carcinoma of the
breast: role of associated lesions and E-cadherin in differential diagnosis. Am J Surg
Pathol 2001;25:237e44.
50. Jacobs TW, Pliss N, Kouria G, et al. Carcinomas in situ of the breast with
indeterminable features: role of E-cadherin staining in categorization. Am J Surg
Pathol 2001;25:229e36.
51. Dabbs DJ, Bhargava R, Chivukula M. Lobular versus ductal breast neoplasms. The
diagnostic utility of p120 catenin. Am J Surg Pathol 2007;31:427e37.
52. Bratthauer GL, Moinfar F, Stamatakos MD, et al. Combined E-cadherin and high
molecular weight cytokeratin immuno-profile differentiates lobular, ductal and hybrid
mammary intraepithelial neoplasias. Hum Pathol 2002;33:620e7.
53. Bratthauer GL, Miettinen M, Tavassoli FA. Cytokeratin immunoreactivity in lobular
intraepithelial neoplasia. J Histochem Cytochem 2003;51:1527e31.
54. Droufakou S, Deshmane V, Roylance R, et al. Multiple ways of silencing E-cadherin
gene expression in lobular carcinoma of the breast. Int J Cancer 2001;92:404e8.
55. Sarrio D, Moreno-Bueno G, Hardisson D, et al. Epigenetic and genetic alterations of
APC and CDH1 genes in lobular breast cancer: relationships with abnormal
E-cadherin and catenin expression and microsatellite instability. Int J Cancer
2003;106:208e15.
56. Mastracci TL, Tjan S, Bane AL, et al. E-cadherin alterations in atypical lobular
hyperplasia and lobular carcinoma in situ of the breast. Mod Pathol
2005;18:741e51.
57. Lu YJ, Osin P, Lakhani S, et al. Comparative genomic hybridization analysis of
lobular carcinoma in situ and atypical lobular hyperplasia and potential roles for
gains and losses of genetic material in breast neoplasia. Cancer Res
1998;58:4721e7.
58. Shelley Hwang E, Nyante SJ, Yi Chen Y, et al. Clonality of lobular carcinoma in
situ and synchronous invasive lobular carcinoma. Cancer 2004;100:2562e72.
59. Morandi L, Marucci G, Foschini MP, et al. Genetic similarities and differences
between lobular in situ neoplasia (LN) and invasive lobular carcinoma of the breast.
Virchows Arch 2006;449:14e23.
60. Page DL, Dupont WD, Rogers LW, et al. Atypical hyperplastic lesions of the female
breast. A long term follow-up study. Cancer 1985;55:2698e708.
61. Page DL, Dupont WD, Rogers LW. Ductal involvement by cells of atypical lobular
hyperplasia in the breast: a long term follow-up study of cancer risk. Hum Pathol
1988;19:201e7.
62. Page DL, Schuyler PA, Dupont WA, et al. Atypical lobular hyperplasia as
a unilateral predictor of breast cancer risk: a retrospective cohort study. Lancet
2003;361:125e9.
63. Ottesen GL, Graversen HP, Blichert-Toft M, et al. Lobular carcinoma in situ of the
female breast. Short term results of a prospective nationwide study. The Danish
Breast Cancer Cooperative Group. Am J Surg Pathol 1993;17:14e21.
64. Chuba PJ, Hamre MR, Yap J, et al. Bilateral risk for subsequent breast cancer after
lobular carcinoma-in-situ: analysis of surveillance, epidemiology and end results
data. J Clin Oncol 2005;23:5534e41.
65. Rosen PP, Kosloff C, Lieberman PH, et al. Lobular carcinoma in situ of the breast:
detailed analysis of 99 patients with average follow-up of 24 years. Am J Surg
Pathol 1978;2:225e51.
66. Collins LC, Baer JH, Tamimi RM, et al. Magnitude and laterality of breast
cancer risk according to histologic type of atypical hyperplasia. Cancer
2007;109:180e7.
67. Li CI, Malone KE, Saltzman BS, et al. Risk of invasive breast carcinoma among
women diagnosed with ductal carcinoma in situ and lobular carcinoma in situ,
1988-2001. Cancer 2006;106:2104e12.
J Clin Pathol 2012;65:771–785. doi:10.1136/jclinpath-2012-200733 783
Review
Downloaded from jcp.bmj.com on September 17, 2012 - Published by group.bmj.com
68. Abner AL, Connolly JL, Recht A, et al. The relation between the presence and
extent of lobular carcinoma in situ and the risk of local recurrence for patients with
infiltrating carcinoma of the breast treated with conservative surgery and radiation
therapy. Cancer 2000;88:1072e7.
69. Fisher ER, Land SR, Fisher B, et al. Pathologic findings from the National Surgical
Adjuvant Breast and Bowel Project. Twelve-year observations concerning lobular
carcinoma in situ. Cancer 2004;100:238e44.
70. Ben-David MA, Kleer CG, Paramagui C, et al. Is lobular carcinoma in situ
as a component of breast carcinoma a risk factor for local failure after
breast-conserving therapy? Results of a matched pair analysis. Cancer
2006;106:28e34.
71. Adepoju LJ, Symmans WF, Babiera GV, et al. Impact of concurrent proliferative
high-risk lesions on the risk of ipsilateral breast carcinoma recurrence and
contralateral breast carcinoma development in patients with ductal carcinoma in
situ treated with breast-conserving therapy. Cancer 2006;106:42e50.
72. Pinder SE, Provenzano E, Reis-Filho JS. Lobular in situ hyperplasia and columnar
cell lesions: diagnosis in breast core biopsies and implications for management.
Pathology 2007;39:208e16.
73. Reis-Filho JS, Pinder SE. Non-operative breast pathology: lobular neoplasia. J Clin
Pathol 2007;60:1321e7.
74. Lakhani SR, Audretsch W, Cleton-Jensen AM, et al. The management of lobular
carcinoma in situ (LCIS). Is LCIS the same as ductal carcinoma in situ (DCIS)? Eur J
Cancer 2006;42:2205e11.
75. Menon S, Porter GJ, Evans AJ, et al. The significance of lobular neoplasia on
needle core biopsy of the breast. Virchows Arch 2008;452:473e9.
76. O’Driscoll D, Britton P, Bobrow L, et al. Lobular carcinoma in situ diagnosed on
core biopsy e what is the clinical significance? Clin Radiol 2001;56:216e20.
77. Cangiarella J, Guth A, Axelrod D, et al. Is surgical excision necessary for the
management of atypical lobular hyperplasia and lobular carcinoma in situ diagnosed
on core needle biopsy? A report of 38 cases and review of the literature. Arch
Pathol Lab Med 2008;132:979e83.
78. Elsheikh TM, Silverman JF. Follow-up surgical excision is indicated when breast
core needle biopsies show atypical lobular hyperplasia or lobular carcinoma in situ.
Am J Surg Pathol 2005;29:534e43.
79. Mahoney MC, Robinson-Smith TM, Shaughnessy EA. Lobular neoplasia at
11- gauge vacuum-assisted stereotactic biopsy: correlation with surgical
excision biopsy and mammographic follow-up. AJR Am J Roentgenol
2006;187:949e54.
80. Houssami N, Caitto S, Bilous M, et al. Borderline breast core needle histology:
predictive values for malignancy in lesions of uncertain malignant potential (B3).
Br J Cancer 2007;96:1253e7.
81. Londero V, Zuiani C, Linda A, et al. Lobular neoplasia: core needle breast biopsy
underestimation of malignancy in relation to radiologic and pathologic features.
Breast 2008;17:623e30.
82. Hwang H, Barke LD, Mendelson EB, et al. Atypical lobular hyperplasia and classic
lobular carcinoma in situ in core biopsy specimens; routine excision is not
necessary. Mod Pathol 2008;21:1208e16.
83. Chivukula M, Haynick DM, Brufsky A, et al. Pleomorphic lobular carcinoma in situ
(PLCIS) on breast core needle biopsies: clinical significance and immunoprofile. Am
J Surg Pathol 2008;32:1721e6.
84. Pickren JW. Significance of occult metastases. A study of breast cancer. Cancer
1961;14:1266e71.
85. Weaver DL. Sentinel lymph nodes and breast carcinoma. Which micrometastases
are significant? Am J Surg Pathol 2003;27:842e5.
86. Huvos AG, Hutter R, Berg JW. Significance of axillary macrometastases and
micrometastases in mammary carcinoma. Ann Surg 1971;173:44e6.
87. Fisher ER, Palekar R, Rockette H, et al. Pathological findings for the National
Surgical Adjuvant Breast and Bowel Project (Protocol # 4). Significance of axillary
nodal micro- and macrometastases. Cancer 1978;42:3032e8.
88. Rosen PP, Saigo P, Braun DW Jr, et al. Occult axillary lymph node
metastases in breast cancer: prognostic significance of tumor size. Ann Surg
1981;194:585e91.
89. Fleming ID, Cooper JS, Henson DE, et al, eds. AJCC Cancer Staging Manual. 5th
edn. Philadelphia: Lippincott-Raven, 1997.
90. Greene FL, Page DL, Fleming ID, et al. AJCC Cancer Staging Manual. 6th edn. New
York: Springer-Verlag, 2002.
91. Sobin LH, Wittekind C, eds. UICC TNM Classification of Malignant Tumours. 6th
edn. New York: John Wiley & Sons, Inc, 2002.
92. Singletary SE, Greene FL, Sobin LH. Classification of isolated tumor cells:
clarification of the 6th edition of the American Joint Committee on Cancer Staging
manual. Cancer 2003;98:2740e1.
93. Connolly JL. Changes and problematic areas in interpretation of the AJCC Cancer
Staging Manual, 6th edition, for breast cancer. Arch Pathol Lab Med
2006;130:287e91.
94. Cserni G, Bianchi S, Boecker W, et al; for the European Working Group for Breast
Screening Pathology. Improving the reproducibility of diagnosing micrometastases
and isolated tumor cells. Cancer 2005;103:358e67.
95. Cserni G, Sapino A, Decker T. Discriminating between micrometastases and
isolated tumor cells in a regional and institutional setting. Breast 2006;15:347e54.
96. Turner RR, Weaver DL, Cserni G, et al. Nodal stage classification for breast
carcinoma: improving interobserver reproducibility through standardized histologic
criteria and image-based training. J Clin Oncol 2008;26:258e63.
97. Cserni G, Bianchi S, Vezzosi V, et al. Variations in sentinel node isolated tumour
cells/micrometastasis and non-sentinel node involvement rates according to
different interpretations of the TNM definitions. Eur J Cancer 2008;44:2185e91.
98. Lyman GH, Guiliano AE, Somerfield MK, et al. American Society of Clinical
Oncology guideline recommendations for sentinel lymph node biopsy in early stage
breast cancer. J Clin Oncol 2005;23:7703e20.
99. Ellis IO, Pinder SE, Bobrow L, et al. Pathology reporting of Breast Disease. Sheffield,
UK: NHS cancer Screening Programmes, 2005.
100. Cserni G. A model for determining the optimum histology of sentinel lymph nodes
in breast cancer. J Clin Pathol 2004;57:467e71.
101. Cserni G, Amendoeira I, Apostolikas N, et al; European Working Group for Breast
Screening Pathology. Pathological work-up of sentinel lymph nodes in breast cancer.
Review of current data to be considered for the formulation of guidelines. Eur J
Cancer 2003;39:1654e67.
102. Turner RR, Ollila D, Stern S, et al. Optimal histopathologic examination of the
sentinel lymph node for breast carcinoma staging. Am J Surg Pathol
1999;23:263e7.
103. Dowlatshahi K, Fan M, Anderson JM, et al. Occult metastases in sentinel nodes
of 200 patients with operable breast cancer. Ann Surg Oncol 2001;8:675e81.
104. Cserni G. Complete sectioning of axillary sentinel nodes in patients with breast
cancer. Analysis of two different step sectioning and immunohistochemistry
protocols in 246 patients. J Clin Pathol 2002;55:926e31.
105. Klevesath MB, Bobrow LG, Pinder SE, et al. The value of immuno-histochemistry in
sentinel lymph node histopathology in breast cancer. Br J Cancer 2005;92:2201e5.
106. Roberts CA, Beitsch PD, Litz CE, et al. Interpretative disparity among pathologists
in breast cancer sentinel lymph node evaluation. Am J Surg 2003;186:324e9.
107. Moore KH, Thaler HT, Tan LK, et al. Immunohistochemically detected tumor cells in
the sentinel lymph nodes of patients with breast carcinoma. Biologic metastasis or
procedural artefact? Cancer 2004;100:929e34.
108. Manzotti M, Dell Orto P, Maisonneuve P, et al. Reverse transcriptase e
polymerase chain reaction assay for multiple mRNA markers in the detection of
breast cancer metastases in sentinel lymph nodes. Int J Cancer 2001;95:307e12.
109. Viale G, Dell’Orto P, Biasi DO, et al. Comparative evaluation of an extensive
histopathologic examination and a real-time reverse transcriptase-polymerase chain
reaction assay for mammoglobin and cytokeratin 19 on axillary sentinel lymph nodes
of breast carcinoma patients. Ann Surg 2008;247:136e42.
110. Martin Martinez MD, Veys I, Majjaj S, et al. Clinical validation of a molecular
assay for intra-operative detection of metastases in breast sentinel lymph nodes.
Eur J Surg Oncol 2009;35:387e92.
111. Mansel RE, Goyal A, Douglas-Jones A, et al. Detection of breast cancer metastasis
in sentinel lymph nodes using intra-operative real time GeneSearch BLN assay in
the operating room: results of the Cardiff study. Breast Cancer Res Treat
2009;115:595e600.
112. Cserni G, Gregori D, Merletti F, et al. Meta-analysis of non-sentinel node
metastases associated with micrometastatic sentinel nodes in breast cancer. Br J
Surg 2004;91:1245e52.
113. Viale G, Maiorano E, Mazzarol G, et al. Histologic detection and clinical implications
of micrometastases in axillary sentinel lymph nodes for patients with breast
carcinoma. Cancer 2001;92:1378e84.
114. den Bakker MA, van Weeszenberg A, de Kanter AY, et al. Non-sentinel lymph
node involvement in patients with breast cancer and sentinel node micrometastasis;
too early to abandon axillary clearance. J Clin Pathol 2002;55:932e5.
115. Dabbs DJ, Fung D, Landsittel D, et al. Sentinel lymph node micrometastasis as
a predictor of axillary tumor burden. Breast J 2004;10:101e5.
116. Carcoforo P, Bergossi L, Basaglia E, et al. Prognostic and therapeutic impact of
sentinel node micrometastasis in patients with invasive breast cancer. Tumori
2002;88:54e5.
117. Cserni G, Bianchi S, Vezzosi V, et al. Sentinel lymph node biopsy in staging small
(upto 15mm) breast carcinomas. Results from a European multi-institutional study.
Pathol Oncol Res 2007;13:5e14.
118. Houvenaeghel G, Nos C, Mignotte H, et al. Micrometastases in sentinel lymph
node in a multicentric study: predictive factors of nonsentinel lymph node
involvement: Groupe de Chirurgiens de la Federation des Centres de Lutte Contre le
Cancer. J Clin Oncol 2006;24:1814e22.
119. Van Rijk MC, Peterse JL, Nieweg OE, et al. Additional axillary metastases and
stage migration in breast cancer patients with micrometastases or
submicrometastases in sentinel lymph nodes. Cancer 2006;107:467e71.
120. Van Deurzen CH, Van HR, Hobbelink MG, et al. Predictive value of tumor load in
breast cancer sentinel lymph nodes for second echelon lymph node metastases.
Cell Oncol 2007;29:497e505.
121. Calhoun KE, Hansen NM, Turner RR, et al. Non-sentinel node metastases in breast
cancer patients with isolated tumor cells in the sentinel node: implications for
completion axillary node dissection. Am J Surg 2005;190:588e91.
122. Millis RR, Springall R, Lee AH, et al. Occult lymph node metastases are of no
prognostic significance in breast cancer. Br J Cancer 2002;86:396e41.
123. Kuijt GP, Voogd AC, van de Poll-Frause LV, et al. The prognostic significance of
axillary lymph node micrometastases in breast cancer patients. Eur J Surg Oncol
2005;31:500e5.
124. Maibenco DC, Dombi GW, Kau TY, et al. Significance of micrometastases on the
survival of women with T1 breast cancer. Cancer 2006;107:1234e9.
125. Chen SL, Hoehne FM, Giuliano AE. The prognostic significance of micrometastases in
breast cancer: a SEER population-based analysis. Ann Surg Oncol 2007;15:3378e84.
784 J Clin Pathol 2012;65:771–785. doi:10.1136/jclinpath-2012-200733
Review
Downloaded from jcp.bmj.com on September 17, 2012 - Published by group.bmj.com
126. Tan LK, Giri D, Hummer AJ, et al. Occult axillary node metastases in breast cancer
are prognostically significant: results in 368 node-negative patients with 20-year
follow-up. J Clin Oncol 2008;26:1803e9.
127. Kahn HJ, Hanna WM, Chapman JA, et al. Biological significance of occult
micrometastases in histologically negative axillary lymph nodes in breast cancer
patients using the recent American Joint Committee on Cancer staging system.
Breast J 2006;12:294e301.
128. Yaziji H, Gown AM, Sneige N. Detection of stromal invasion in breast cancer: the
myoepithelial markers. Adv Anat Pathol 2000;7:100e9.
129. Foschini MP, Scarpellini F, Gown AM, et al. Differential expression of moyepithelial
markers in salivary, sweat and mammary glands. Int J Surg Pathol 2000;8:29e37.
130. Lazard D, Sastre X, Frid MG, et al. Expression of smooth muscle specific proteins in
myoepithelium and stromal myofibroblasts of normal and malignant human breast
tissue. Proc Natl Acad Sci U S A 1993;90:9990e1003.
131. Dairkee SH, Blayney C, Smith HS, et al. Monoclonal antibody that defines human
myoepithelium. Proc Natl Acad Sci U S A 1985;82:7409e13.
132. Nagle RB, Bocker W, David JR, et al. Characterization of breast carcinomas by two
monoclonal antibodies distinguishing myoepithelial from luminal epithelial cells.
J Histochem Cytochem 1986;34:869e81.
133. Otterbach F, Bankfalvi A, Bergher S, et al. Cytokeratin 5/6 immunohistochemistry
assists the differential diagnosis of atypical proliferations of the breast.
Histopathology 2000;37:232e40.
134. Joshi MG, Lee AK, Pedersen CA, et al. The role of immunocytochemical markers in
the differential diagnosis of proliferative and neoplastic lesions of the breast. Mod
Pathol 1996;9:57e62.
135. Dwarakanath S, Lee AK, Delellis RA, et al. S100 protein positivity in breast
carcinomas: a potential pitfall in diagnostic immunohistochemistry. Hum Pathol
1987;18:1144e8.
136. Gusterson BA, Monaghan P, Manhendran R, et al. Identification of myoepithelial
cells in human and rat breasts by anti-common acute lymphoblastic leukaemia
antigen antibody A12. J Natl Cancer Inst 1986;77:343e9.
137. Moritani S, Kushima R, Sugihara H, et al. Availability of CD10
immunohistochemistry as a marker of breast myoepithelial cells on paraffin
sections. Mod Pathol 2002;15:397e405.
138. Kovacs A, Walker RA. P-cadherin as a marker in the differential diagnosis of breast
lesions. J Clin Pathol 2003;546:139e41.
139. Barbareschi M, Pecciarini L, Cangi MG, et al. p63, a p53 homologue, is a selective
nuclear marker of myoepithelial cells of the human breast. Am J Surg Pathol
2001;25:1056e60.
140. Werling RW, Hwang H, Yaziji H, et al. Immunohistochemical distinction of invasive
from non-invasive breast lesions. A comparative study of p63 versus calponin and
smooth muscle myosin heavy chain. Am J Surg Pathol 2003;27:82e90.
141. Simpson PT, Gale T, Reis-Filho JS, et al. Distribution and significance of 14-3-3
sigma, a novel moyepithelial marker, in normal, benign and malignant tissue.
J Pathol 2004;202:274e85.
142. Reis-Filho JS, Milanezi F, Silva P, et al. Maspin expression in myoepithelial
tumours of the breast. Pathol Res Pract 2002;197:817e21.
143. Douglas-Jones A, Shah V, Morgan J, et al. Observer variability in the
histopathological reporting of core biopsies of papillary breast lesions is reduced by
the use of immunohistochemistry for CK5/6, calponin and p63. Histopathology
2005;47:202e8.
144. Collins LC, Carlo VP, Hwang H, et al. Intracystic papillary carcinomas of the breast:
a re-evaluation using a panel of myoepithelial cell markers. Am J Surg Pathol
2006;30:1002e7.
145. Tse GM, Tan PH, Lui PC, et al. The role of immunohistochemistry for smooth
muscle actin, p63, CD10 and CK14 in the differential diagnosis of papillary lesions of
the breast. J Clin Pathol 2007;60:315e20.
146. Tan PH, Aw MY, Yip G, et al. Cytokeratins in papillary lesions of the breast. Is there
a role in distinguishing intraductal papilloma from papillary ductal carcinoma in situ.
Am J Surg Pathol 2005;29:625e32.
147. Rabban JT, Koerner FC, Lerwill MF. Solid papillary ductal carcinoma in situ versus
usual ductal hyperplasia in the breast: a potentially difficult distinction resolved by
cytokeratin 5/6. Hum Pathol 2006;37:787e93.
148. Ribeiro-Silva A, Zamzelli Ramalho LN, Garcia SB, et al. Is p63 reliable in detecting
microinvasion in ductal carcinoma in situ of the breast. Pathol Oncol Res
2003;9:20e3.
149. Kalof AN, Tam D, Beatty B, et al. Immunostaining patterns of myoepithelial cells in
breast lesions: a comparison of CD10 and smooth muscle myosin heavy chain. J
Clin Pathol 2004;57:625e9.
150. Hungermann D, Buerger H, Oehlschregel C, et al. Adenomyoepithelial tumours
and myoepithelial carcinomas of the breast e a spectrum of monophasic and
biphasic tumours dominated by immature myoepithelial cells. BMC Cancer
2005;5:92e102.
151. Turner NC, Reis-Filho JS. Basal-like breast cancer and the BRCA1 phenotype.
Oncogene 2006;25:5846e53.
152. Banerjee S, Reis-Filho JS, Ashley S, et al. Basal-like breast carcinomas: clinical
outcome and response to chemotherapy. J Clin Pathol 2006;59:729e35.
153. Neilsen TO, Hsu FD, Jensen K, et al. Immunohistochemical and clinical
characterization of the basal-like subtype of invasive breast carcinoma. Clin Cancer
Res 2004;10:5367e74.
154. Fulford LG, Easton DP, Reis-Filho JS, et al. Specific morphological features
predictive for the basal phenotype in grade 3 invasive ductal carcinoma of breast.
Histopathology 2006;49:22e34.
155. Arnes JB, Brunet JS, Stefannson I, et al. Placental cadherin and the basal
epithelial phenotype of BRCA-1 related breast cancer. Clin Cancer Res
2005;11:4003e11.
156. Rakha EA, El-Sayed ME, Green AR, et al. Breast carcinoma with basal
differentiation: a proposal for pathology definition based on basal cytokeratin
expression. Histopathology 2007;50:434e8.
157. Fisher B, Dignam J, Wolmark N, et al. Tamoxifen in treatment of intraductal breast
cancer: National Surgical Adjuvant Breast and Bowel Project B-24 randomised
controlled trial. Lancet 1999;353:1993e2000.
158. Fisher B, Land S, Mamounas E, et al. Prevention of invasive breast cancer in
women with ductal carcinoma in situ: an update of the National Surgical Adjuvant
Breast and Bowel Project experience. Semin Oncol 2001;28:400e18.
159. Allred DC, Bryant J, Land S, et al. Estrogen receptor expression as a predictive
marker of the effectiveness of tamoxifen in the treatment of DCIS: findings from the
NSABP Protocol B-24 (abstract 30). Br Cancer Res Tr 2002;76:S36.
160. Houghton J, George WD, Cusick J, et al. Radiotherapy and tamoxifen in women
with completely excised ductal carcinoma in situ of the breast in the UK, Australia
and New Zealand: randomised controlled trial. Lancet 2003;362:95e192.
161. Fisher B, Constantino JP, Wickerham L, et al. Tamoxifen for prevention of breast
cancer: report of the National Surgical Adjuvant Breast and Bowel Project P-1 study.
J Natl Cancer Inst 1998;90:1371e88.
162. Anon. Pathology Reporting of Breast Disease. NHS Breast Screening Programme
Publication No 58. NHS Cancer Screening Programmes and the Royal College of
Pathologists. 2005:87.
163. Livasy CA, Perou CM, Karaca G, et al. Identification of a basal-like subtype of
breast ductal carcinoma in situ. Hum Pathol 2007;38:197e204.
164. Leong AS, Sormunen RT, Vinyuvat S, et al. Biologic markers in ductal carcinoma in
situ and concurrent infiltrating carcinoma. A comparison of eight contemporary
grading systems. Am J Clin Pathol 2001;115:709e18.
165. McCarty KS Jr, Szabo E, Flowers JL, et al. Use of a monoclonal anti-estrogen
receptor antibody in the immunohistochemical evaluation of human tumors. Cancer
Res 1986;46(8 Suppl):4244se8s.
166. Harvey JM, Clark GM, Osborne CK, et al. Estrogen receptor status by
immunohistochemistry is superior to the ligand binding assay for predicting
response to adjuvant endocrine therapy in breast cancer. J Clin Oncol
1999;17:1474e81.
167. Thomas JS, Hanby AM, Pinder SE, et al; on behalf of the Sloane Project Steering
Group. Implications of inconsistent measurement of ER status in non-invasive breast
cancer: a study of 1684 cases from the Sloane Project. Breast J 2008;14:31e6.
168. Baqai T, Sousha S. Oestrogen receptor negativity as a marker for high grade ductal
carcinoma in situ of the breast. Histopathology 2003;42:440e7.
169. Otteson GL, Christesen IJ, Larsen JK, et al. Carcinoma in situ of the breast:
correlation of hstopathology to immunohstochemical markers and DNA ploidy.
Breast Cancer Res Treat 2000;60:219e26.
170. Bryan BB, Schnitt SJ, Collins LC. Ductal carcinoma in situ with basal-like
phenotype: a possible precursor to invasive basal-like breast cancer. Mod Pathol
2006;19:617e21.
171. Dabbs DJ, Chivukula M, Carter G, et al. Basal phenotype of ductal carcinoma in
situ: recognition and immunohistologic profile. Mod Pathol 2006;19:1506e11.
PAGE fraction trail=15
J Clin Pathol 2012;65:771–785. doi:10.1136/jclinpath-2012-200733 785
Review
Downloaded from jcp.bmj.com on September 17, 2012 - Published by group.bmj.com
doi: 10.1136/jclinpath-2012-200733
J Clin Pathol 2012 65: 771-785 originally published online July 19, 2012
Rosemary A Walker, Andy Hanby, Sarah E Pinder, et al.
pathology
Current issues in diagnostic breast
http://jcp.bmj.com/content/65/9/771.full.html
Updated information and services can be found at:
These include:
References
http://jcp.bmj.com/content/65/9/771.full.html#ref-list-1
This article cites 160 articles, 30 of which can be accessed free at:
service
Email alerting
the box at the top right corner of the online article.
Receive free email alerts when new articles cite this article. Sign up in
Collections
Topic
• Immunology (including allergy) (1325 articles)
• Editor's choice (71 articles)
Articles on similar topics can be found in the following collections
Notes
http://group.bmj.com/group/rights-licensing/permissions
To request permissions go to:
http://journals.bmj.com/cgi/reprintform
To order reprints go to:
http://group.bmj.com/subscribe/
To subscribe to BMJ go to:
Downloaded from jcp.bmj.com on September 17, 2012 - Published by group.bmj.com